DK2288700T3 - HBV EPITOP-REACTIVE EXOGEN T-CELL RECEPTOR (TCR) AND APPLICATIONS THEREOF - Google Patents

HBV EPITOP-REACTIVE EXOGEN T-CELL RECEPTOR (TCR) AND APPLICATIONS THEREOF Download PDF

Info

Publication number
DK2288700T3
DK2288700T3 DK09742949.2T DK09742949T DK2288700T3 DK 2288700 T3 DK2288700 T3 DK 2288700T3 DK 09742949 T DK09742949 T DK 09742949T DK 2288700 T3 DK2288700 T3 DK 2288700T3
Authority
DK
Denmark
Prior art keywords
seq
hbv
chain
tcr
epitope
Prior art date
Application number
DK09742949.2T
Other languages
Danish (da)
Inventor
Antonio Bertoletti
Adam Gehring
Original Assignee
Agency Science Tech & Res
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Agency Science Tech & Res filed Critical Agency Science Tech & Res
Application granted granted Critical
Publication of DK2288700T3 publication Critical patent/DK2288700T3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/29Hepatitis virus
    • A61K39/292Serum hepatitis virus, hepatitis B virus, e.g. Australia antigen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4632T-cell receptors [TCR]; antibody T-cell receptor constructs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/464838Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6425Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a receptor, e.g. CD4, a cell surface antigen, i.e. not a peptide ligand targeting the antigen, or a cell surface determinant, i.e. a part of the surface of a cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • C12Q1/701Specific hybridization probes
    • C12Q1/706Specific hybridization probes for hepatitis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57438Specifically defined cancers of liver, pancreas or kidney
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/53Liver
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/01DNA viruses
    • G01N2333/02Hepadnaviridae, e.g. hepatitis B virus
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/14Heterocyclic carbon compound [i.e., O, S, N, Se, Te, as only ring hetero atom]
    • Y10T436/142222Hetero-O [e.g., ascorbic acid, etc.]
    • Y10T436/143333Saccharide [e.g., DNA, etc.]

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Urology & Nephrology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Pathology (AREA)
  • Biophysics (AREA)
  • Communicable Diseases (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • General Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Hospice & Palliative Care (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)

Description

DESCRIPTION
Field of the invention [0001] The present invention relates generally to the field of immune mediated therapies for treatment of Hepatitis B Virus (HBV) and HBV-related diseases such as cirrhosis and Hepatocellular carcinoma (HCC) and to methods of preparing these therapies. In particular, the immune mediated therapies may be in the form of cells expressing at least one HBV epitope-reactive exogenous T cell receptor and/or polynucleotides, vectors and/or polypeptides encoding the HBV epitope-reactive T cell receptor.
Background of the art [0002] Hepatitis B virus (HBV) infects the liver of hominoidae, including humans, and causes an inflammation called hepatitis. It is a DNA virus and one of many unrelated viruses that cause viral hepatitis. HBV currently infects at least 350 million people worldwide. 75% of chronically infected HBV patients are living in Asia but the virus has a world-wide diffusion. For example, in the United States, approximately 1.5 million Americans, or about 0.5% of the population have HBV. The infection is especially more common in certain risk groups like men who have sex with other men, renal dialysis patients and persons with haemophilia. Chronic HBV infections affects 10 to 15% of first generation Asian Americans and approximately 5% of children adopted from Russia, Asia and Eastern Europe have chronic HBV infections. Chronic HBV infections may eventually lead to liver cirrhosis and Hepatocellular carcinoma (HCC), a fatal disease with very poor response to current chemotherapy.
[0003] Currently available methods of treatment for this global large reservoir of chronically infected subjects are challenging as existing drugs suppress but do not eliminate HBV. Though majority of HBV infected patients respond to currently available methods of treatment showing improvements in liver histology and serum Alanine transaminase (ALT) levels, almost all patients relapse when treatment is stopped. Furthermore, the more common methods of treatment of HBV involve the use of drugs such as lamivudine and adefovir. These drugs however, result in development of anti-viral resistance in the patients, which occur in approximately 20% of patients treated with lamivudine and in about 3% of those treated with adefovir each year. Ultimately a large proportion of patients would develop resistance, at which point, the anti-viral drugs would have little effect.
[0004] There is thus a global need for an effective method of anti-viral therapy that results in an HBV specific immune response to efficiently and successfully eliminate the covalently closed circular form of HBV in a patient. Naturally occurring HBV-epitope specific T cells found in some subjects, lead to the subjects having an efficient and innate control of the HBV infection. Past studies have shown that patients with persistent/chronic HBV infection have their HBV-epitope specific T cells deleted or functionally altered. Therefore, increased knowledge of virus-host interactions during HBV infection has prompted speculation that therapeutic restoration of the defective anti-viral immunity present in patients with chronic infection could possibly lead to disease resolution. The validity of this concept was directly demonstrated in patients with chronic HBV infection who underwent bone marrow transplantation and received marrows from donors with natural immunity to HBV. Infusion of a healthy HBV-primed immune system led to resolution of chronic HBV infection in these patients. However, bone marrow transplantation is clearly not an easy therapeutic option for chronically HBV infected patients and attempts to boost HBV-specific immunity using various vaccines in patients with chronic hepatitis B have been disappointing.
[0005] A potential cause of failure of the therapeutic vaccine strategy is the fact that the immune system of HBV chronic carriers does not have the same efficiency and repertoire of specificities as that of healthy non-HBV infected subjects. Further, persistent high production of viral antigens in chronic HBV infected patients can delete or tolerize antigen-specific T cells. Chronic HBV infected patients are thus characterized by low/absent HBV-specific CD4+ and CD8+ T cell responses. Moreover, it has been speculated that both lowT cell avidity and an ineffective cytokine profile generated in response to HBV infection may contribute to the development of chronic HBV infection rather than viral clearance.
[0006] Clearly, more effective therapies targeting molecules involved in the cytokine profile of HBV infected patients are necessary in order to reduce the worldwide morbidity and mortality from HBV infection and HBV-related malignancies.
Summary of the invention [0007] The present invention addresses the problems above, and in particular provides a novel, efficient and effective method of treating HBV infection by redirecting the specificity of lymphocytes of chronic HBV infected patients using exogenous T cell receptor (TCR) transfer.
[0008] According to a first aspect, the present invention provides at least one isolated T cell comprising at least one HBV epitope-reactive exogenous T cell receptor (TCR) and/ or fragment thereof, wherein the HBV epitope is HLA-A2 restricted and is: HBc18-27 epitope comprising SEQ ID NO:25 and/or SEQ ID NO:26; and/or HBs370-79 epitope comprising any one of the sequences selected from the group consisting of SEQ ID NO:56 to 58, and wherein the fragment comprises an active site which enables the HBV-epitope-reactive exogenous T cell receptor to recognise the HBc18-27 and/or the HBs370-79 epitope. In particular, the HBs370-79 epitope may comprise sequence SEQ ID NO:56.
[0009] In particular, the T cell according to the present invention may further comprise at least one second HBV epitope-reactive exogenous TCR and/or fragment thereof, wherein the second HBV epitope may be different from the first HBV epitope. In particular, the first HBV epitope may be HBc18-27 and the second epitope may be HBs370-79.
[0010] The exogenous TCR may comprise at least one α-chain comprising at least one amino acid sequence selected from the group consisting of SEQ ID NO:9, SEQ ID NO:10 and SEQ ID NO:11 or mutants thereof. In particular, the exogenous TCR comprises at least one α-chain comprising the sequences SEQ ID NO:9, SEQ ID NO: 10 and SEQ ID NO:11. The exogenous TCR may comprise at least one β-chain comprising at least one amino acid sequence selected from the group consisting of SEQ ID NO:21, SEQ ID NO:22 and SEQ ID NO:23 or mutants thereof. More in particular, the exogenous TCR may comprise at least one β-chain comprising the sequences SEQ ID NO:21, SEQ ID NO:22 and SEQ ID NO:23. All sequences are shown in Tables 1 and 2.
[0011] In particular, the exogenous TCR may comprise at least one α-chain having at least 90% amino acid identity to SEQ ID NO: 12 or a fragment thereof and/ or at least one β-chain having at least 90% amino acid identity to SEQ ID NO:24 or a fragment thereof, wherein the fragment comprises an active site which enables the exogenous T cell receptor to recognise the HBc18-27 and/or the HBs370-79 epitope. More in particular, the exogenous TCR may comprise at least one α-chain of SEQ ID NO: 12 and/ or at least one β-chain of SEQ ID NO:24.
[0012] The exogenous TCR may comprise at least one α-chain comprising at least one amino acid sequence selected from the group consisting SEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:46 or mutants thereof. In particular, the exogenous TCR comprises at least one α-chain comprising the sequences SEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:46. The exogenous TCR may comprise at least one β-chain comprising at least one amino acid sequence selected from the group consisting of SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:54 or mutants thereof. More in particular, the exogenous TCR may comprise at least one β-chain comprising the sequences SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:54. All sequences are shown in Tables 1 and 2. The mutant retains the ability to bind and activate the HBV epitope.
[0013] In particular, the exogenous TCR may comprise at least one α-chain having at least 90% amino acid identity to SEQ ID NO:47 or a fragment thereof and/ or at least one β-chain having at least 90% amino acid identity to SEQ ID NO:55 or a fragment thereof, wherein the fragment comprises an active site which enables the exogenous T cell receptor to recognise the HBc18-27 and/or the HBs370-79 epitope. More in particular, the exogenous TCR may comprise at least one α-chain of SEQ ID NO:47 and/ or at least one β-chain of SEQ ID NO:55.
[0014] .The T cell of the present invention may be at least one Peripheral Blood Lymphocytes (PBL) -derived T cell.
[0015] According to another aspect, the present invention provides at least one isolated polynucleotide comprising at least one sequence encoding at least one α-chain and/or at least one sequence encoding at least one β-chain wherein the α-chain and β-chain are part of at least one exogenous HBV epitope-reactive TCR, and wherein the HBV epitope is HLA-A2 restricted and is: HBc18-27 epitope comprising SEQ ID NO:25 and/or 26; and/or HBs370-79 epitope comprising any one of the sequences selected from the group consisting of SEQ ID NO: 56 to 58.
[0016] In particular, the sequence encoding the α-chain comprises at least one sequence selected from SEQ ID NO:1 and SEQ ID NO:5, at least one sequence selected from SEQ ID NO:2 and SEQ ID NO:6 and at least one sequence selected from SEQ ID NO:3 and SEQ ID NO:7 and/or the sequence encoding the β-chain comprises at least one sequence selected from SEQ ID NO:13 and SEQ ID NO: 17, at least one sequence selected from SEQ ID NO: 14 and SEQ ID NO: 18 and at least one sequence selected from SEQ ID NO:15 and SEQ ID NO:19. More in particular, the sequence encoding the α-chain may comprise SEQ ID NO:5, SEQ ID NO:6 and SEQ ID NO:7 and/or the sequence encoding the β-chain may comprise SEQ ID NO: 17, SEQ ID NO: 18 and SEQ ID NO: 19.
[0017] In particular, the α-chain of the HBV epitope-reactive exogenous TCR may have at least 80% sequence identity to SEQ ID NO:4 or SEQ ID NO:8 and/or the β-chain of the HBV epitope-reactive exogenous TCR may have at least 80% sequence identity to SEQ ID NO: 16 or SEQ ID NO:20. More in particular, the sequence encoding the α-chain may be selected from the group consisting of SEQ ID NO:4 and SEQ ID NO:8, and/or the sequence encoding the β-chain may be selected from the group consisting of SEQ ID NO:16 and SEQ ID NO:20. More in particular, the sequence of the α-chain of the HBc18-27 epitope reactive exogenous TCR may be selected from the group consisting of SEQ ID NO:4 and SEQ ID NO:8, and/or the sequence of the β-chain of the HBc18-27 epitope reactive exogenous TCR may be selected from the group consisting of SEQ ID NO:16 and SEQ ID NO:20.
[0018] In particular, the sequence encoding the α-chain comprises SEQ ID NO:40, SEQ ID NO:41 and SEQ ID NO:42 and/or the sequence encoding the β-chain comprises SEQ ID NO:48, SEQ ID NO:49, and SEQ ID NO:50. More in particular, the α-chain of the HBV epitope-reactive exogenous TCR may have at least 80% sequence identity to SEQ ID NO:43 and the β-chain of the HBV epitope-reactive exogenous T cell receptor has at least 80% sequence identity to SEQ ID NO:51. Even more in particular, the sequence encoding the α-chain comprises SEQ ID NO:43, and/or the sequence encoding the β-chain comprises SEQ ID NO:51. More in particular, the sequence of the α-chain of the HBs370-79 epitope reactive exogenous TCR may comprise SEQ ID NO:43 and/or the sequence of the β-chain of the HBs370-79 epitope reactive exogenous TCR may comprise SEQ ID NO:51.
[0019] According to another aspect, the present invention provides at least one isolated polypeptide encoded by at least one polynucleotide of the present invention.
[0020] The isolated polypeptide comprises: (a) an α-chain comprising SEQ ID NO:9, SEQ ID NO:10 and SEQ ID NO:11 and/or a β-chain comprising SEQ ID NO:21, SEQ ID NO:22 and SEQ ID NO:23; and/or (b) an α-chain comprisingSEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:46 and/or a β-chain comprising SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:54; wherein the polypeptide is at least one HBV epitope-reactive exogenous T cell receptor and the HBV epitope is HLA-A2 restricted.
[0021] In particular, the α-chain may have at least 90% amino acid identity to SEQ ID NO: 12 and the β-chain may have at least 90% amino acid identity to SEQ ID NO:24. In particular, the α-chain may have at least 90% amino acid identity to SEQ ID NO:47 and the β-chain may have at least 90% amino acid identity to SEQ ID NO:55. In particular, the HBV epitope may be HBc18-27, HBs370-79 and/or a mutant thereof. More in particular, the polypeptide may be at least one soluble TCR or a fragment thereof. Even more in particular, the soluble TCR or fragment thereof may be linked to at least one anti-viral drug.
[0022] The present disclosureprovides at least one construct comprising the polynucleotide of the present invention operably connected to at least one promoter.
[0023] The present disclosurefurther provides at least one vector comprising the construct according to the present invention or the polynucleotide according to the present invention.
[0024] The present disclosurealso provides at least one T cell comprising the vector or the construct according to the present invention.
[0025] The present disclosurealso provides at least one method of preparing at least one T cell comprising at least one HBV epitope-reactive exogenous TCR for delivery to at least one subject the method comprising transducing at least one T cell isolated from the subject with the construct of or the vector of the present invention.
[0026] The present disclosurefurther provides at least one method of preparing at least one HBV epitope-reactive exogenous TCR comprising: 1. (a) isolating at least one HBV-epitope reactive T cell from at least one HBV-exposed individual that resolved HBV infection; 2. (b) cloning at least one polynucleotide sequence encoding at least one a-and/or β-chain of at least one TCR cell receptor from the HBV-epitope reactive T cell of step (a); 3. (c) delivering the polynucleotide sequence of step (b) to at least one cell; and 4. (d) incubating the cell under conditions suitable for expression of the HBV epitope-reactive exogenous T cell receptor by the cell.
[0027] In particular, the method of preparing at least one HBV epitope-reactive exogenous TCR comprises the isolation of at least one T cell, step (a) which may be HBc18-27 epitope reactive and/or HBs370-79 epitope reactive.
[0028] According to one aspect, the present invention provides at least one T cell according to the present invention and/or a polynucleotide according to the present invention and/or a polypeptide according to the present invention, for use in the treatment of HBV infection and/or HBV-related hepatocellular carcinoma.
[0029] The present disclosureadditionally provides at least one method of treating HBV and/or inhibiting reactivation of HBV in at least one subject comprising administering to the subject at least one immunotherapeutically effective amount of cells, at least one vector and/or at least one polypeptide according to any aspect of the present invention.
[0030] The present disclosurealso provides at least one method of treating HBV-related hepatocellular carcinoma in at least one subject comprising administering to the subject at least one immunotherapeutically effective amount of cells, at least one vector and/or at least one polypeptide according to any aspect of the present invention [0031] According to one aspect, the present invention provides at least one in vitro method for diagnosing at least one subject that is able to resolve HBV infection, the method comprising: 1. (a) providing at least one sample from at least one subject; 2. (b) detecting the presence of at least one polynucleotide comprising, substantially consisting of or consisting of at least one nucleic acid sequence selected from SEQ ID NO: 4, SEQ ID NO: 8, SEQ ID NO: 16, SEQ ID NO: 20, SEQ ID NO: 43, SEQ ID NO: 51, and/or a homologue thereof; and/or 3. (c) detecting the presence of at least one polypeptide comprising, substantially consisting of or consisting of the amino acid sequence of SEQ ID NO: 12, SEQ ID NO: 24, SEQ ID NO: 47, SEQ ID NO: 55, and/or a homologue thereof; wherein the presence of the polynucleotide and/or the polypeptide is indicative of the subject being able to resolve the HBV infection.
[0032] According to yet another aspect, the present invention provides at least one in vitro method for diagnosing at least one subject as having or as being at risk of having HBV infections and/or HBV related hepatocellular carcinoma, the method comprising: 1. (a) providing at least one sample from at least one subject; 2. (b) detecting the presence of at least one polynucleotide comprising, substantially consisting of or consisting of at least one nucleic acid sequence selected from SEQ ID NO: 4, SEQ ID NO: 8, SEQ ID NO: 16, SEQ ID NO: 20, SEQ ID NO: 43, SEQ ID NO: 51, a homologue and/or a fragment thereof; and/or 3. (c) detecting the presence of at least one polypeptide comprising, substantially consisting of or consisting of the amino acid sequence of SEQ ID NO: 12, SEQ ID NO: 24, SEQ ID NO: 47, SEQ ID NO: 55, a homologue and/or a fragment thereof; wherein the absence of the polynucleotide and/or the polypeptide correlates with the likelihood of the subject as having or as being at risk for having HBV-infection and/or HBV-related hepatocellular carcinoma.
[0033] According to another aspect, the present disclosurefurther provides at least one use of at least one cell, at least one vector and/or at least one polypeptide according to any aspect of the present invention in the preparation of a medicament for treating HBV infection and/or HBV-related hepatocellular carcinoma.
[0034] The present disclosurealso provides at least one kit for detecting and/or treatment of HBV infection and/or HBV-related hepatocellular carcinoma, the kit comprising at least one cell, at least one vector and/or at least one polypeptide according to any aspect of the present invention.
Brief description of the figures [0035]
Figure 1 shows a schematic diagram of the steps involved in producing a cell comprising at least one HBV epitope-reactive exogenous T cell receptor. It comprises the isolation of a T cell clone from an HLA-A2 positive HBV patient who has resolved the infection, using the T cell clone to form a retroviral vector construct encoding HBV T cell receptor and using the vector to form cells that express the HBV epitope-reactive exogenous T cell receptor Figure 1 was partly taken from Schumacher et al., 2002.
Figure 2 (A) depicts FACS analysis of peripheral blood lymphocytes (PBL) which were stimulated with HBc18-27 peptide to generate T cell lines. The HBc18-27 specific T cell line was isolated using specific HBc18-27-HLA-A201 pentamers and magnetic separation. The isolated HBc18-27 specific T cell line was stained with HBc18-27-HLA-A201 specific pentamers to confirm clonality. The results show that 100% of the T cells were CD8+ and HLA-A2 pentamer positive, indicating clonality.
Figure 2 (B) shows a graph with the results of V beta typing of a potential T cell clone, C183, which was derived from a resolved HLA-A201 HBV patient using staining with a panel of TCR V beta monoclonal antibodies (MAbs). MAbs reacting with the human TCR V beta region are almost as specific as a private idiotypic MAb in identifying T cell clones. This method was thus used to evaluate expression of V beta gene families. The clone, C183, which was found to be highly specific to HBV core 18-27 (HBc18-27) epitope. The results suggest that all T cells were νβ8 positive.
Figure 3 shows the cytotoxic function of HBc18-27 specific Cytotoxic T lymphocyte (CTL) clone C183. CarboxyFluoroscein Succinimidyl Ester (CFSE) labelled HepG2 cells pulsed with HBc18-27 (8-10 M) epitope were mixed with HepG2 cells that were not HBc18-27 pulsed. The HepG2 cells were cultured in the presence or absence of CTL clones at a 1:1 ratio for 5 h. Cytotoxicity was determined by disappearance of CFSE labelled targets in the presence of C183 clone (+C183). After 5 h co-incubation, the results show that only CFSE negative cells (no HBc18-27 peptide) remained and nearly all HBc18-27 positive, CFSE positive HepG2 cells were eliminated by C183, indicating the T cell clone is capable of killing tumour cells presenting HBV antigens like HBc18-27. There was no significant difference in the cell count when no CTL was added (NO CTL).
Figure 4 shows that C183 T cell clone recognizes endogenously processed viral antigen presented by hepatocellular carcinoma cells.
Figure 4 (A) shows the results of HBc18-27 specific Cytotoxic T lymphocyte (CTL), C183, incubated with parental, vector only, HepG2 cell line (HepG2TA2-7) for 5 h as a control. The control was tested for cytokine release and cytotoxic degranulation (CD107a+ staining), which revealed that C183 did not respond to the control cell line HepG2TA2-7 as there was no activation to degranulate (CD107a+) and did not produce IFN-y.
Figure 4 (B) shows that C183 T cell clone activation after co-culture with HepG2.105 cells which expressed different levels of the HBV antigen stimulated IFN-γ production and degranulation, indicating the C183 T cell clone recognizes the HBc18-27 epitope processed and presented by hepatocellular carcinoma cells. 84% of T cell clones were activated after 5 h co-culture with HepG2 cells expressing HBV.
Figure 5 shows the results of how C183 T cell clone responds to naturally infected hepatocytes. Primary hepatocytes which were isolated from HLA-A2+ or HLA-A2- explanted chronic HBV livers were co-cultured with C183. HBc18-27-specific C183 T cell clone degranulation and IFN-γ production was measured after incubation of C183 alone, with HLA-A2+ uninfected, with HLA-mismatch HBV infected or with HLA-A2+ HBV infected primary human hepatocytes. C183 activation (12% CD107a+) was only observed following incubation with HLA-A2+ HBV infected primary human hepatocytes indicating that the C183 T cell clone recognizes endogenously processed viral antigen presented by HBV infected primary hepatocytes that express the correct HLA molecules on their cell surface.
Figure 6 shows the ability of T cell clone, C183, to recognize the HBc18-27 epitope from HBV genotypes B and C (grey bars, Asia virus; C18-I) and HBV genotypes A and D (black bars, European/American virus; C18-V) presented by different subtypes of the HLA-A2 MHC class I family. Epstein-Barr virus (EBV) transformed B cells with known subtypes of HLA-A2 were loaded with increasing concentrations of HBc18-27 epitope from different genotypes of HBV and co-cultured with C183 T cell clone for 5 h. IFN-γ production was measured to determine T cell activation. Data shows that (A) HLA-A201; (B) HLA-A206; and (C) HLA-A207 MHC class I molecules could present the HBV epitope from all genotypes and, were recognized by C183 and thus stimulated efficient IFN-γ production by the T cells. C183 T cell clone was found to be able to recognize the HBc18-27 epitope with high sensitivity (s 1 pM) from genotypes B and C or genotypes A and D and the epitope was recognized almost equally when presented by the three most dominant HLA-A2 subtypes A0201 (A), A0206 (B) and A0207 (C).
Figure 7 shows the analysis of T cell Receptor (TCR) transduction efficiency.
Figure 7 (A) shows the FACS analysis of peripheral blood lymphocytes (PBL) when they were mock transduced (negative control) or transduced with the HBc18-27 A201 TCR comprised of νβ8.2 and Va3 chains. PBL were stained for νβ8 chain 3 days after transduction to determine expression of the introduced HBc18-27 TCR. Mock transduced PBL expressed endogenous levels of νβ8 (CD8+=1.8%; CD4+=2.1). Expression of νβ8 increased substantially after transduction with HBc18-27 A201 TCR compared to mock transduced cells, which expressed only endogenous levels of νβ8, indicating successful transduction and expression.
Figure 7 (B) shows the results obtained when mock or HBc18-27 TCR transduced cells were stained with HBc18-27-A201 specific pentamer to determine if the Va3 chain was expressed and properly paired with νβ8.2 chain on the cell surface. Pentamer analysis showed that only HBc18-27 TCR transduced T cells expressed the appropriate a and β chains recognized by the HBc18-27-A201 specific pentamer and that the a and β chains were properly paired on the cell surface. No staining was detected in Mock transduced cells because PBL came from a healthy donor which was HLA-A2 negative.
Figure 7 (C) shows the mean frequency of νβ8+ T cells from 5 healthy, 5 HBeAg+ chronic HBV patients (HBV DIMA > 107 copies/ml), and 5 HBeAg-chronic HBV patients (HBV DNA< 106 copies/ml). This indicates that the transduced νβ8 TCR chain is expressed equally well in chronic HBV patients and healthy donors.
Figure 7 (D) shows the mean frequency of HBc18-27-A201 specific pentamer+ T cells from 5 healthy, 5 HBeAg+ chronic HBV patients (HBV DIMA > 107 copies/ml), and 5 HBeAg- chronic HBV patients (HBV DIMA < 106 copies/ml). This indicates that the properly paired transduced TCR is expressed equally well in chronic HBV patients and healthy donors.
Figure 8 (A) shows the FACS analysis of T cells transduced with HBc18-27 A201 TCR to confirm that the introduced HBc18-27 TCR was functional. HBc18-27 TCR transduced or mock transduced T cells were co-cultured overnight with HLA-A2+ T2 cells which were first pulsed with HBc18-27 peptide ("peptide") and incubated for 5h. The function of the T cells was then assessed by intracellular cytokine staining for IFN-γ (left column), TNF-α (middle column) and IL-2 (right column). In the absence of peptide, HBc18-27 TCR transduced cells were not activated (top row). Mock transduced cells were not activated by peptide pulsed T2 cells (middle row). HBc18-27 TCR transduced cells were stimulated to produce IFIM-y, TNF-α, and IL-2 following co-culture with peptide pulsed T2 cells (bottom row). Both CD8+ and CD8- (CD4+ T cells) were capable of producing all three cytokines confirming function of the introduced TCR in CD4+ T cells and multifunctional capacity of TCR re-directed cells.
Figure 8 (B) shows that HBc18-27 TCR transduced T cells from chronic HBV patients are equally functional to healthy donors. Mean frequency of IFIM-y, TNF-α and IL-2 positive cells from 5 healthy, 5 HBeAg+ chronic HBV patients (HBV DIMA > 107 copies/ml), and 5 HBeAg- chronic HBV patients (HBV DIMA< 106 copies/ml) are shown. The results show that T cells transduced with HBc18-27-A201 TCR become multifunctional.
Figure 9 shows the affinity of HBc18-27 TCR transduced T cells from healthy and chronic HBV patients compared to original C183 T cell clone.
Figure 9 (A) shows the results when IFN-y+ CD8+ HBc18-27 TCR transduced T cells and C183 T cell clones were co-cultured with T2 cells pulsed with increasing concentrations of HBc18-27 peptide for 5 h and then stained for IFN-γ. Data is presented as mean percentage of maximum response for HBc18-27 TCR transduced T cells derived from 5 healthy donors, 5 HBeAg+ chronic HBV patients (HBV DIMA > 107 copies/ml), and 5 HBeAg- chronic HBV patients (HBV DIMA < 106 copies/ml) where C183 is 100% specific while TCR transduced cell frequency varies. Results show that the affinity for the viral epitope of CD8+ HBc18-27 TCR transduced T cells derived from patient T cells was identical when compared to the original C183 T cell clone and recognized the epitope down to concentrations of 0.1 - 1 pg/ml.
Figure 9 (B) shows the results when IFN-y+ CD4+ HBc18-27 TCR transduced T cells and C183 T cell clones were co-cultured with T2 cells pulsed with increasing concentrations of HBc18-27 peptide for 5 h and then stained for IFN-γ Data is presented as mean percentage of maximum response for HBc18-27 TCR transduced T cells derived from 5 healthy donors, 5 HBeAg+ chronic HBV patients (HBV DIMA > 107 copies/ml), and 5 HBeAg- chronic HBV patients (HBV DIMA <10® copies/ml) where C183 is 100% specific while TCR transduced cell frequency varies. CD4+HBc18-27 TCR transduced cells derived from patient T cells were less sensitive compared to C183 but 50% of HBc18-27 specific T cells were activated at 100 pg/ml indicating that the introduced HBc18-27 TCR was highly sensitive, even in the absence of the CD8 co-receptor.
Figure 10 shows the expression of optimized HBc18-Opt-TCR compared to the wild type HBc18-WT-TCR.
Figure 10 (A) shows histograms with the percentage of νβ8+ T cells from mock, HBc18-WT-TCR and HBc18-Opt-TCR transduced PBL cells. Frequency of νβ8+ cells was comparable between the optimized and wild type HBc18-TCR transduced cells.
Figure 10 (B) shows the comparison of the expression level of νβ8 in mock, HBc18-WT-TCR and HBc18-Opt-TCR transduced T cells. To compare the level of expression, mean fluorescent intensity (MFI) for νβ8+ cells was used. The results show that there was no significant difference in the level of νβ8 expression between optimized and wild type TCR transduced cells.
Figure 10 (C) shows the results obtained from HBc18-27 HLA-A2 specific pentamer staining. Cells from each group mentioned above were stained with anti-CD8 and HBc18-27 HLA-A2 specific pentamers. Nearly a two-fold increase in the frequency of pentamer positive cells in T cells transduced with HBc18-Opt-TCR constructs were shown compared to the wild type TCR suggesting that the Va3 chain may be expressed more efficiently from the codon optimized construct compared to the wild type sequence.
Figure 11 shows that there is an increased frequency of IFIM-γ positive cells in T cells transduced with HBc18-Opt-TCR compared to HBc18-WT-TCR constructs. Mock transduced T cells, HBc18-WT-TCR and HBc18-Opt-TCR were stimulated overnight with HLA-A2 positive T2 cells which were initially pulsed or not pulsed with HBc18-27 peptide, in the presence of 2 pg/ml brefeldin A. T cells were then labelled with anti-CD8 PE-Cy7 and stained for IFIM-y by intracellular cytokine staining. The results demonstrated that there was nearly a two fold increase in the frequency of IFN-γ positive T cells transduced with the HBc18-Opt-TCR compared to T cells transduced with the HBc18-WT-TCR. The increase in pentamer positive cells correlated with an increase in functional cells able to respond to peptide loaded target T cells.
Figure 12 (A) EBV transformed B cells expressing FILA-A0201, -A0206 or-A0207 subtypes were loaded with increasing concentrations of HBV genotype A/D HBc18-27 epitope (sequence provided in Table 1) and co-cultured with HBc-18-27 TCR transduced T cells for 5h. T cell activation was measured by intracellular cytokine staining for IFN-γ. The results show that the HBc-18-27 TCR transduced T cells recognize the HBc18-27 eptiope presented by multiple HLA-A2 subtypes.
Figure 12 (B) EBV transformed B cells expressing HLA-A0201, -A0206 or-A0207 subtypes were loaded with increasing concentrations of HBV genotype B/C HBc18-27 epitope (sequences provided in Table 1) and co-cultured with HBc-18-27 TCR transduced T cells for 5h. T cell activation was measured by intracellular cytokine staining for IFN-γ. The results show that the HBc-18-27 TCR transduced T cells recognize the HBc18-27 eptiope presented by multiple HLA-A2 subtypes.
Figure 12 (C) show the results when HLA-A2+ T2 cells were loaded with each of the mutant peptides of HBc18-27 epitope and co-cultured with HBc-18-27 TCR transduced T cells for 5 h. T cell recognition of the mutant peptides was determined by intracellular cytokine staining for IFN-γ. The sequences of the mutant peptides are provided in Table 1. As can be seen from the results, the HBc-18-27 TCR transduced T cells recognized various mutants of the HBc18-27 eptiope. The Unstim 1 and Unstim 2 refer to the control where the HLA-A2+ T2 cells were not loaded wth any mutant peptide.
Figure 13 (A) TCR transduced T cells from each patient group (healthy, HBeAg(-), HBeAg(+)) were co-cultured with DiOC labeled HepG2 cells expressing the entire HBV genome (HBV-HepG2) or the parental control cell line (Ctrl-HepG2) at EffectonTarget ratio of 1:1 for 6 h in the presence of Propidium Iodide (PI). Dying target T cells were DiOC+/PI+. Results are representative of 3 experiments. Effector T cells were considered to be IFIM-y+/CD8+ cells determined by intracellular cytokine staining after stimulation with peptide pulsed T2 cells. The results showed that HBc18-27 TCR transduced T cells can kill HCC cell lines which endogenously express HBV proteins or are loaded with the HBV 18-27 peptide.
Figure 13 (B) DiOC labeled HCC cell lines (HepG2, SNU-387, SNU-368) were loaded with increasing concentrations of HBc18-27 peptides and co-cultured with HBc-18-27 TCR transduced T cells at Effector to target ratio of 1:1 for 6 h in the presence of Propidium Iodide (PI). Dying target T cells were DiOC+/PI+. Results are representative of 3 experiments. Effector T cells were considered to be IFN-y+/CD8+ cells determined by intracellular cytokine staining after stimulation with peptide pulsed T2 cells. The results showed that HBc18-27 TCR transduced T cells are able to kill multiple HLA-A2+ HCC cell lines.
Figure 14 (A) shows that the E10-1D T cell clone or HBs370-79 TCR transduced T cells, recognize HBs370-79 epitope loaded HLA-A2+ T2 cells. E10-1D T cell clone is made according to the method provided in Example 10. T2 cells were loaded with increasing concentrations of HBs370-79 peptide and co-cultured with E10-1D or HBs370-79 TCR transduced T cells for 5 h and stained for IFN-γ. Data presented as percent maximum response to normalize differing frequency of each line. The results show that HBs370-79 specific TCR is functional.
Figure 14 (B) shows that HBs370-79 TCR transduced T cells recognize genotypic variants of the HBs370-79 epitope (The sequences of the genotypic variants are provided in Table 2). HBs370-79 TCR transduced T cells were co-cultured with HLA-A2+ T2 cells loaded with 1 pg/ml peptide of each genotype for 5 h and activation was assessed by CD107a labeling and staining for IFN-γ. Genotype A &amp; D sequences are identical. The results show that HBs370-79 specific TCR recognizes genotypic variants of the HBs370-79 epitope.
Detailed description of the invention [0036] Bibliographic references mentioned in the present specification are for convenience listed in the form of a list of references and added at the end of the examples.
Definitions [0037] For convenience, certain terms employed in the specification, examples and appended claims are collected here.
[0038] The term "comprising" is herein defined to be that where the various components, ingredients, or steps, can be conjointly employed in practicing the present invention. Accordingly, the term "comprising" encompasses the more restrictive terms "consisting essentially of" and "consisting of."
[0039] With the term "consisting essentially of" it is understood that the exogenous TCR polypeptide and/or polynucleotide according to the invention "substantially" comprises the indicated sequence as "essential" element. Additional sequences may be included at the 5' end and/or at the 3' end. Accordingly, a polypeptide "consisting essentially of" sequence X will be novel in view of a known polypeptide accidentally comprising the sequence X
[0040] With the term "consisting of it is understood that the polypeptide and/or polynucleotide according to the invention corresponds to at least one of the indicated sequence (for example a specific sequence indicated with a SEQ ID Number or a homologous sequence or fragment thereof).
[0041] The term "exogenous T cell receptor" (TCR) is herein defined as a recombinant TCR which is expressed in a cell by introduction of exogenous coding sequences for a TCR. In particular, the HBV epitope-reactive TCR may be expressed in a cell in which the TCR is either not natively expressed or is expressed at levels that are insufficient to induce a response by the cell or a responder cell upon TCR ligand binding.
[0042] The term "fragment" is herein defined as an incomplete or isolated portion of the full sequence of the HBV epitope-reactive exogenous TCR which comprises the active site(s) that confers the sequence with the characteristics and function of the HBV epitope-reactive exogenous TCR. In particular, it may be shorter by at least one nucleotide or amino acid. More in particular, the fragment comprises the active site(s) that enable the HBV epitope-reactive exogenous TCR to recognise the HBc18-27 epitope and/or the HBs370-79 epitope.
[0043] The term "HBV epitope-reactive T cell Receptor (TCR)" is herein defined as a TCR which binds to an HBV epitope in the context of a Major Histocompatibility Complex (MHC) molecule to induce a helper or cytotoxic response in the cell expressing the recombinant TCR. In particular, the HBV epitope may be HBc18-27. More in particular, the HBV epitope may comprise the sequence of SEQ ID NO:25. The HBV epitope may be HBs370-79. More in particular, the HBV epitope may comprise the sequence of SEQ ID NO:56, SEQ ID NO:57 or SEQ ID NO:58.
[0044] The term "HBc 18-27 epitope" is herein defined as an epitope that can stimulate HLA class I restricted T cells. It may be used interchangeably in the present invention as HBc18, HBc18-27, HBc18-27 peptide and the peptide. The sequence of the epitope may be "FLPSDFFPSV" (SEQ ID NO:25). In the present invention, the term HBc18-27 is used to refer to the HBc18-27 epitope of genotype A/D prevalent amongst Caucasians of sequence SEQ ID NO:25 unless otherwise stated. The region of the T cell receptor that binds to the epitope is referred to as HBc18-27 TCR or HBc18 TCR.
[0045] The term "HBs370-79 epitope" is herein defined as an epitope that can stimulate HLA class I restricted T cells. The sequence of the epitope may be "SIVSPFIPLL" (SEQ ID NO:56). In the present invention, the term HBs370-79 is used to refer to the HBs370-79 epitope of genotype A/D prevalent amongst Caucasians of sequence SEQ ID NO:56 unless otherwise stated. The region of the T cell receptor that binds to the epitope is referred to as HBs370-79 TCR.
[0046] The term "immunotherapeutically effective amount" is herein defined as an amount which results in an immune-mediated prophylactic or therapeutic effect in the subject, i.e., that amount which will prevent or reduce symptoms at least 50% compared to pre-treatment symptoms or compared to a suitable control.
[0047] The term "isolated" is herein defined as a biological component (such as a nucleic acid, peptide or protein) that has been substantially separated, produced apart from, or purified away from other biological components in the cell of the organism in which the component naturally occurs, i.e., other chromosomal and extrachromosomal DNAand RNA, and proteins. Nucleic acids, peptides and proteins which have been isolated thus include nucleic acids and proteins purified by standard purification methods. The term also embraces nucleic acids, peptides and proteins prepared by recombinant expression in a hosT cell as well as chemically synthesized nucleic acids.
[0048] The term "operably connected" herein defined as a functional linkage between regulatory sequences (such as a promoter and/or array of transcription factor binding sites) and a second nucleic acid sequence, wherein the regulatory sequences direct transcription of the nucleic acid corresponding to the second sequence.
[0049] The term "mutant" or "mutant form" of a TCR epitope is herein defined as one which has at least one amino acid sequence that varies from at least one reference virus-encoded sequence via substitution, deletion or addition of at least one amino acid, but retains the ability to bind and activate the TCR bound and activated by the non-mutated epitope. In particular, the mutants may be naturally occurring or may be recombinantly or synthetically produced.
[0050] The term "soluble TCR" is herein defined as a soluble (secreted) form of the membrane bound TCR which is the molecule that is responsible for the T cell's recognition of the antigen for which it is specific. In its soluble form it is analogous to a monoclonal antibody except that it recognizes fragments of peptides associated with MHC molecules while antibodies recognize determinants on a whole protein. In particular, the isolated polypeptide of the present invention may be used for producing a soluble TCR using any method known in the art. More in particular, the soluble TCR may bind to the HBc18-27 and/or HBs370-79 epitope of HBV.
[0051] The term "subject" is herein defined as vertebrate, particularly mammal, more particularly human. For purposes of research, the subject may particularly be at least one animal model, e.g., a mouse, rat and the like. In particular, for the animal models, the sequence of the TCR a- and β- chains may be selected based on species. In some cases, transgenic animals expressing human MHC molecules may also be useful in evaluating specific aspects of the present invention.
[0052] A person skilled in the art will appreciate that the present invention may be practised without undue experimentation according to the method given herein. The methods, techniques and chemicals are as described in the references given or from protocols in standard biotechnology and molecular biology text books.
[0053] In one aspect of the present invention, there is provided at least one isolated T cell comprising at least one HBV epitope-reactive exogenous T cell receptor (TCR) and/ or fragment thereof, wherein the HBV epitope is HLA-A2 restricted and is: HBc18-27 epitope comprising SEQ ID NO:25 and/or SEQ ID NO:26; and/or HBs370-79 epitope comprising any one of the sequences selected from the group consisting of SEQ ID NO:56 to 58, and wherein the fragment comprises an active site which enables the HBV-epitope-reactive exogenous T cell receptor to recognise the HBc18-27 and/or the HBs370-79 epitope. Such T cells may be suitable for use in adoptive transfer protocols to provide a particularly effective mode of treatment. The isolated T cells of the present invention may circumvent the problem of deletion and sub-optimal function of HBV-specific CD8+ and CD4+ cells which may be present in patients with chronic HBV infection. In particular, the TCR from a patient who resolved the HBV infection may be used to redirect the specificity of the lymphocytes of chronic HBV patients for TCR transfer.
[0054] In particular, HBV-reactive TCRs may be prepared by transforming or transducing at least one isolated T cell with one or more polynucleotides encoding functional a- and/or β- chains and/or polypeptides of functional a- and/or β- chains that may assemble to form at least one functional HBV-epitope reactive TCR. In particular, the isolated T cell may be isolated using any technique known in the art. More in particular, at least one cell isolation kit, Ficoll-Paque density gradient centrifugation, BD FACSAria Cell Sorting System and the like may be used to isolate the T cell.
[0055] The isolated T cell may be at least one autologous cell, i.e., they may be derived from at least one subject that may receive the resultant transduced or transformed cells. In particular, the isolated T cells may be derived from the peripheral blood lymphocytes and/or hematopoietic stem cells of the subject. More in particular, the isolated cell may be at least one T cell that innately expresses at least one CD4+ cell surface marker, at least one CD8+ cell surface marker, both CD4+ and CD8+ marker or neither CD4+ nor CD8+ cell surface markers. In particular, the cell according to the present invention may be at least one T cell that also natively expresses at least one TCR. The HBV-reactive exogenous TCR may bind the same epitope as the natively expressed TCR, and/or may bind a different epitope. In particular, the HBV-reactive exogenous TCR may be expressed in at least one T cell in which the TCR may be either not natively expressed or may be expressed at levels that may be insufficient to induce response by the T cell or responder cell upon TCR ligand binding. In particular, the isolated T cell according to the present invention may be transduced with two or more different HBV-reactive exogenous TCRs, i.e., TCRs which bind to two or more different HBV epitopes.
[0056] HBV-exposed patients who have cleared their viral infections, may provide excellent source of HBV-reactive T cells expressing high affinity TCRs. In particular, HBV-epitope reactive TCRs may be prepared by isolating at least one HBV-reactive T cell from at least one HBV-exposed aviremic individual, and cloning the polynucleotide sequence encoding the a- and β-chains of the TCR from the HBV reactive T cell. Once these sequences have been cloned using standard methods known in the art, the sequences may be delivered to at least one isolated cell and the isolated cell may be incubated under conditions suitable for expression of the TCR by the isolated cell. More in particular, the suitable conditions may include standard cell culture conditions. When the TCR is expressed in vitro or ex vivo, the cell expressing the TCR may be evaluated for reactivity with HBV epitopes, among other parameters of interest, as known in the art and as exemplified below. In some protocols, i.e., those wherein the vector may be administered to at least one subject, the TCR may also be expressed in vivo to provide a therapeutic effect in at least one subject in need thereof, i.e., at least one subject with acute or chronic HBV infection or HBV-associated condition.
[0057] The HBV-reactive TCRs according to the present invention may be functional in the isolated cell in which they may be expressed. In particular, they may be functional heterodimers of a and β TCR chains associated with a CD3 complex that recognizes at least one HBV epitope in the context of at least one Class I or Class II MHC molecule. In humans, the MHC restriction of at least one epitope may be dependent on at least one particular Human Leukocyte Antigen (HLA) expressed by at least one cell presenting the antigen. HBV-reactive TCRs that may recognize HBV epitopes may be restricted to a HLA type (i.e., HLA-A, HLA-B, HLA-C, HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQB1, HLA-DRA, and HLA- DRB1). For purposes of study, the HBV-reactive TCR may recognize at least one HBV epitope in the context of at least one MHC molecule of at least one species other than human, e.g., H-2Kof mouse.
[0058] The HBV-reactive TCR recognizes HBV epitopes that are HLA-A2 restricted. Approximately 50% of the general population expresses the MHC class I molecule HLA-A2, an HLA-A serotype. Therefore, HLA-A2-restricted TCRs may find widespread therapeutic use. In particular, the subtype may identify gene products of many HLA-A*02 alleles, comprising HLA-A*0201, *0202, *0203, *0206, and *0207 gene products. There may be distinct differences in the subtypes between Caucasian and Asian populations. Whereas more than 95% of the HLA-A2 positive Caucasian population is HLA-A0201 the HLA-A2 positive Chinese population may be broken down into 23% HLA-A0201; 45% HLA-A0207; 8% HLA-A0206; 23% HLA-A0203.
[0059] The TCRs of the present invention are HBV-epitope reactive. A list of known immunoreactive HBV epitopes and their sequences may be found on page 233, chapter 11 (The effect of pathogens on the immune system: Viral hepatitis) of the book In immunodominance: The choice of the Immune System, J.A. Frelinger, ed (Weinheim: Wiley-VCH). The HBV epitope may comprise at least one core antigen, envelope antigen, surface antigen and/or mutants thereof.
[0060] The HBV epitope comprises at least one hepatitis B core antigen, and/or mutants thereof. More in particular, the HBV epitope may comprise HBc18-27, and/or mutants thereof. The HBc18-27 epitope may comprise "FLPSDFFPSV" (SEQ ID NO:25) or mutants thereof. Furthermore, the HBc18-27 epitope present in genotype A and D which may comprise "FLPSDFFPSV (SEQ ID NO: 25), which are most prevalent in Europe, differs from HBc18-27 epitope of genotypes B and C which may comprise "FLPSDFFPSr (SEQ ID NO: 26), which are most prevalent in Asia.
[0061] In particular, the HBV epitope may comprise at least one hepatitis B envelope antigen, and/or mutants thereof. More in particular, the HBV epitope may comprise HBs370-79, and/or mutants thereof. The HBs370-79 epitope may comprise "SIVSPFIPLL" (SEQ ID NO:56) or mutants thereof. Furthermore, the HBs370-79 epitope present in genotype Aand D which may comprise "SIVSPFIPLL" (SEQ ID NO:56), which are most prevalent in Europe, differs from HBs370-79 epitope of genotype B which may comprise "NILSPFMPLL" (SEQ ID NO: 57), and epitope of genotype C which may comprise "NILNPFLPLL" (SEQ ID NO: 58), which are most prevalent in Asia.
[0062] In particular, the T cell according to the present invention may further comprise at least one second HBV epitope-reactive exogenous TCR and/or fragment thereof, wherein the second HBV-reactive exogenous TCR may be the same epitope as the first epitope or different from the first HBV epitope. In particular, the second HBV epitope may be HBs370-79 and the first epitope may be HBc18-27.
[0063] The exogenous TCR may comprise at least one α-chain comprising at least one amino acid sequence selected from the group consisting the sequences SEQ ID NO:9, SEQ ID NO:10 and SEQ ID NO:11 and/or mutants thereof. In particular, the exogenous TCR comprises at least one α-chain comprising the sequences SEQ ID NO:9, SEQ ID NO: 10 and SEQ ID NO:11. More in particular, the α-chain comprises all three sequences, SEQ ID NO:9, SEQ ID NO:10 and SEQ ID NO:11. The α-chain may comprise the sequences of SEQ ID NO:9, SEQ ID NO: 10 and SEQ ID NO: 11 consecutively. In particular, the α-chain may consist essentially of the sequences SEQ ID NO:9, SEQ ID NO:10 and SEQ ID NO:11. All sequences used in the present invention are given in Tables 1 and 2.
[0064] The exogenous TCR may further comprise at least one β-chain comprising at least one amino acid sequence selected from the group consisting of the sequences SEQ ID NO:21, SEQ ID NO:22 and SEQ ID NO:23 and/or mutants thereof. In particular, the β-chain may comprise all three sequences, SEQ ID NO:21, SEQ ID NO:22 and SEQ ID NO:23. In particular, the β-chain may comprise the sequences of SEQ ID NO:21, SEQ ID NO:22 and SEQ ID NO:23 consecutively. More in particular, the β-chain may consist essentially of the sequences SEQ ID NO:21, SEQ ID NO:22 and SEQ ID NO:23.
[0065] In particular, the exogenous TCR may comprise at least one α-chain having at least 80% amino acid identity to SEQ ID NO: 12 or a fragment thereof and/ or at least one β-chain having at least 80% amino acid identity to SEQ ID NO:24 or a fragment thereof. In particular, the α-chain and β-chain may be at least about 85%, at least about 90%, or at least about 95% identical to SEQ ID NO:12 and SEQ ID NO:24 respectively. More in particular, the α-chain may comprise all sequences SEQ ID NO:9, SEQ ID NO: 10 and SEQ ID NO:11 and β-chain may comprise all sequences SEQ ID NO:21, SEQ ID NO:22 and SEQ ID NO:23 [0066] The exogenous TCR may comprise at least one α-chain comprising at least one amino acid sequence selected from the group consisting the sequences SEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:46 and/or mutants thereof. In particular, the exogenous TCR comprises at least one α-chain comprising the sequences SEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:46. More in particular, the α-chain comprises all three sequences, SEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:46. The a-chain may comprise the sequences of SEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:46 consecutively. In particular, the α-chain may consist essentially of the sequences SEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:46. All sequences used in the present invention are given in Tables 1 and 2.
[0067] The exogenous TCR may further comprise at least one β-chain comprising at least one amino acid sequence selected from the group consisting of the sequences SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:54 and/or mutants thereof. In particular, the β-chain may comprise all three sequences, SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:54. In particular, the β-chain may comprise the sequences of SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:54 consecutively. More in particular, the β-chain may consist essentially of the sequences SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:54.
[0068] In particular, the exogenous TCR may comprise at least one α-chain having at least 80% amino acid identity to SEQ ID NO:47 or a fragment thereof and/ or at least one β-chain having at least 80% amino acid identity to SEQ ID NO:55 or a fragment thereof. In particular, the α-chain and β-chain may be at least about 85%, at least about 90%, or at least about 95% identical to SEQ ID NO:47 and SEQ ID NO:55 respectively. More in particular, the α-chain may comprise all sequences SEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:46 and β-chain may comprise all sequences SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:54.
[0069] The sequences encoding the α-chain and β-chain have been determined to be the amino acid sequence for at least one productively rearranged α-chain and β-chain respectively of a TCR reactive against HBV epitope HBc18-27 and/or HBs370-79.
[0070] More in particular, the exogenous TCR reactive against HBV epitope HBc18-27 may comprise at least one α-chain of SEQ ID NO:12 and/ or at least one β-chain of SEQ ID NO:24. Even more in particular, the exogenous TCR reactive against HBV epitope HBc18-27 may consists essentially of at least one α-chain of SEQ ID NO:12 and at least one β-chain of SEQ ID NO:24. The exogenous TCR reactive against HBV epitope HBc18-27 may consists of at least one α-chain of SEQ ID NO:12 and at least one β-chain of SEQ ID NO:24. In particular, the exogenous TCR reactive against HBV epitope HBc18-27 may consist of two a-chains and two β-chains.
[0071] More in particular, the exogenous TCR reactive against HBV epitope HBs370-79 may comprise at least one α-chain of SEQ ID NO:47 and/ or at least one β-chain of SEQ ID NO:55. Even more in particular, the exogenous TCR reactive against HBV epitope HBs370-79 may consists essentially of at least one α-chain of SEQ ID NO:47 and at least one β-chain of SEQ ID NO:55. The exogenous TCR reactive against HBV epitope HBs370-79 may consists of at least one α-chain of SEQ ID NO:47 and at least one β-chain of SEQ ID NO:55. In particular, the exogenous TCR reactive against HBV epitope HBs370-79 may consist of two a-chains and two β-chains.
[0072] Percentage identity may be determined using the algorithm of Karlin and Altschul (Proc. Natl. Acad. Sci. 87: 2264-68 (1990), modified Proc. Natl. Acad. Sci. 90: 5873-77 (1993)). Such algorithm is incorporated into the BLASTx program, which may be used to obtain amino acid sequences homologous to a reference polypeptide. The present invention may also encompass TCR a- or β-chains having amino acid sequences including conservative amino acid substitutions. Such substitutions are well known in the art.
[0073] According to another aspect, the present invention provides at least one isolated polynucleotide comprising at least one sequence encoding at least one α-chain and/or at least one sequence encoding at least one β-chain wherein, the encoded a-chain and β-chain may be part of at least one HBV epitope-reactive TCR, wherein the HBV epitope is HLA-A2 restricted and is: HBc18-27 epitope comprising SEQ ID NO:25 and/or SEQ ID NO:26; and/or HBs370-79 epitope comprising any one of the sequences selected from the group consisting of SEQ ID NO: 56 to 58.
[0074] In particular, the sequence encoding the α-chain comprises at least one sequence selected from SEQ ID NO:1 and SEQ ID NO:5, at least one sequence selected from SEQ ID NO:2 and SEQ ID NO:6 and at least one sequence selected from SEQ ID NO:3 and SEQ ID NO:7 and/or the sequence encoding the β-chain comprises at least one sequence selected from SEQ ID NO:13 and SEQ ID NO: 17, at least one sequence selected from SEQ ID NO: 14 and SEQ ID NO: 18 and at least one sequence selected from SEQ ID NO:15 and SEQ ID NO:19. More in particular, the sequence encoding the α-chain may comprise SEQ ID NO:5, SEQ ID NO:6 and SEQ ID NO:7 and/or the sequence encoding the β-chain may comprise SEQ ID NO: 17, SEQ ID NO: 18 and SEQ ID NO: 19.
[0075] In particular, the sequence encoding the α-chain of the HBc-18-27 epitope-reactive TCR may have at least 80% sequence identity to SEQ ID NO:4 or SEQ ID NO:8 and/or the sequence encoding the β-chain of the HBc-18-27 epitope-reactive exogenous TCR may have at least 80% sequence identity to SEQ ID NO: 16 or SEQ ID NO:20. In particular, the sequence(s) encoding the α-chain and β-chain may be at least about 85%, at least about 90%, or at least about 95% identical to SEQ ID NO:4 or SEQ ID NO:8 and SEQ ID NO: 16 or SEQ ID NO:20 respectively. More in particular, the sequence encoding the α-chain may be selected from the group consisting of SEQ ID NO:4 and SEQ ID NO:8, and/or the sequence encoding the β-chain may be selected from the group consisting of SEQ ID NO:16 and SEQ ID NO:20. The HBc-18-27 epitope-reactive exogenous TCR may comprise two α-chains encoded by the sequence SEQ ID NO:8 and two β-chains encoded by SEQ ID NO:20. More in particular, the a-chain and β-chain may be encoded by nucleotide sequences that comprise the consecutive sequence shown in SEQ ID NO:4 or SEQ ID NO:8 and SEQ ID NO: 16 or SEQ ID NO:20 respectively.
[0076] Even more in particular, the exogenous TCR may consists essentially of at least one α-chain encoded by SEQ ID NO:4 or SEQ ID NO:8 and at least one β-chain encoded by SEQ ID NO:16 or SEQ ID NO:20. The exogenous TCR may consist of at least one α-chain encoded by SEQ ID NO:4 or SEQ ID NO:8 and at least one β-chain encoded by SEQ ID NO: 16 or SEQ ID NO:20. In particular, the exogenous TCR may consist of two α-chains encoded by SEQ ID NO:4 or SEQ ID NO:8 and two β-chains encoded by SEQ ID NO:16 or SEQ ID NO:20.
[0077] In particular, the sequence encoding the α-chain comprises SEQ ID NO:40, SEQ ID NO:41 and SEQ ID NO:42 and/or the sequence encoding the β-chain comprises SEQ ID NO:48, SEQ ID NO:49, and SEQ ID NO:50.
[0078] More in particular, the sequence encoding the α-chain of the HBs370-79 epitope-reactive exogenous TCR may have at least 80% sequence identity to SEQ ID NO:43 and the β-chain of the HBs370-79 epitope-reactive exogenous T cell receptor has at least 80% sequence identity to SEQ ID NO:51. In particular, the sequence(s) encoding the α-chain and β-chain may be at least about 85%, at least about 90%, or at least about 95% identical to SEQ ID NO:43 and/or SEQ ID NO:51 respectively. More in particular, the sequence encoding the α-chain may be SEQ ID NO:43 and SEQ ID NO:51, and/or the sequence encoding the β-chain may be SEQ ID NO:51. The HBs370-79 epitope-reactive exogenous TCR may comprise two α-chains encoded by the sequence SEQ ID NO:43 and two β-chains encoded by SEQ ID NO:51. More in particular, the α-chain and β-chain may be encoded by nucleotide sequences that comprise the consecutive sequence shown in SEQ ID NO:43 and SEQ ID NO:51 respectively.
[0079] The present invention further provides at least one isolated polypeptide encoded by the polynucleotides of the present invention.
[0080] The at least one isolated polypeptide may comprise at least one amino acid sequence selected from the group consisting of SEQ ID NO:9, SEQ ID NO: 10 and SEQ ID NO:11. In particular, the polypeptide may have at least 80% amino acid identity to SEQ ID NO: 12. More in particular, the polypeptide may be at least about 85%, at least about 90%, or at least about 95% identical to SEQ ID NO:12.
[0081] The at least one isolated polypeptide may comprise at least one sequence selected from the group consisting of SEQ ID NO:44, SEQ ID NO:45 and SEQ ID NO:46. In particular, the polypeptide may have at least 80% amino acid identity to SEQ ID NO:47. More in particular, the polypeptide may be at least about 85%, at least about 90%, or at least about 95% identical to SEQ ID NO:47.
[0082] The polypeptide may further comprise at least one sequence selected from the group consisting of SEQ ID NO:21, SEQ ID NO:22 and SEQ ID NO:23. In particular, the further sequence may have at least 80% amino acid identity to SEQ ID NO:24. More in particular, the further sequence may be at least about 85%, at least about 90%, or at least about 95% identical to SEQ ID NO:24. Even more in particular, the polypeptide may comprise the consecutive sequence of amino acids shown in SEQ ID NO:12 and SEQ ID NO:24 respectively.
[0083] The polypeptide may further comprise at least one sequence selected from the group consisting of SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:54. In particular, the further sequence may have at least 80% amino acid identity to SEQ ID NO:55. More in particular, the further sequence may be at least about 85%, at least about 90%, or at least about 95% identical to SEQ ID NO:55. Even more in particular, the polypeptide may comprise the consecutive sequence of amino acids shown in SEQ ID NO:47 and SEQ ID NO: 55 respectively.
[0084] The polypeptide according to the present invention may be at least one HBV epitope-reactive exogenous TCR. In particular, the polypeptide may be functionally equivalent to a specifically exemplified TCR sequence that may have been modified by single or multiple amino acid substitutions, by addition and/or deletion of amino acids, or where one or more amino acids have been chemically modified, but which nevertheless retains the binding specificity and high affinity binding activity of the TCR protein of the present invention to the HBV epitope. In particular, the HBV epitope may be HBc18-27, HBs370-79 or a mutant thereof. More in particular, the polypeptide may be at least one soluble TCR. Even more in particular, the soluble TCR protein may lack the portions of the native cell-bound TCR and may be stable in solution (i. e., it does not generally aggregate in solution when handled as described herein and under standard conditions for protein solutions).
[0085] The soluble TCR may be prepared by any method known in the art. Examples of processes that may be used to prepare the soluble TCR may comprise but are not limited to constructing polymeric receptor chains in which an immunoglobulin heavy chain variable region from at least one phosphorylcholine-specific antibody may be substituted with TCR a and β variable regions, introducing translational termination codons upstream of the TCR transmembrane region or replacing the transmembrane domains of the TCR a and β chain cDNAs with a signal for glycosylphosphatidyl inositol (GPI) linkage from the carboxy terminus of the GPI linked protein Thy-1.
[0086] The soluble TCR may be linked to at least one anti-viral drug. The anti-viral drug may target HBV. For example, anti-viral drugs may comprise, but are not limited to, adefovir dipivoxil, interferon alfa-2b, pegylated interferon alfa-2a, lamivudine, entecavir, telbivudine and the like.
[0087] In particular, the soluble TCR may be linked to the anti-viral drugs to form soluble TCR-drug conjugates by any means known in the art. The link may be a chemical moiety comprising a covalent bond or a chain of atoms that covalently attaches a soluble TCR to a drug moiety. Linkers may comprise a divalent radical such as an alkylene, an arylene, a heteroarylene, moieties such as:-(CR2)nO(CR2)n- ; repeating units of alkyloxy (e.g. polyethylenoxy, PEG, polymethyleneoxy) and alkylamino (e.g. polyethyleneamino, Jeffamine™); and diacid ester and amides including succinate, succinamide, diglycolate, malonate, caproamide and the like.
[0088] The present disclosure provides at least one construct comprising the polynucleotide of the present disdosureoperably connected to at least one promoter. The coding sequences for a- and β- chains of the TCR may be operably connected to at least one promoter functional in the isolated cell. Suitable promoters may be constitutive and inducible promoters, and the selection of an appropriate promoter may be well within the skill in the art. For example, suitable promoters may comprise, but are not limited to, the retroviral LTR, the SV40 promoter, the CMV promoter and cellular promoters (e.g., the β-actin promoter).
[0089] The present disclosurealso provides at least one vector comprising the construct according to the present disclosureor the polynucleotide according to the present invention. In particular, the vectors may comprise, but not limited to, lentiviral vectors, retroviral vectors, adenoviral vectors, adeno-associated virus vectors and Herpes Simplex Virus vectors. More in particular, retroviral vectors may be used for delivery of the constructs either in vitro, ex vivo or in vivo, as described in the examples.
[0090] The present disclosurealso provides at least one T cell comprising the vector, the construct or the polypeptide according to the presentdisdosure and invention.
[0091] The present disdosurefurther provides at least one method of preparing at least one T cell comprising at least one HBV epitope-reactive exogenous TCR for delivery to at least one subject comprising transducing at least one T cell isolated from the subject with the construct of and/or the vector of the present disclosure. Constructs and vectors according to the present disclosuremay be delivered to cells in vitro, ex vivo or in vivo using any number of methods known to those of skill in the art. For example, if the cells are in vitro or ex vivo, they may be transformed or transduced according to standard protocols, e.g., those described in Molecular Cloning: A Laboratory Manual, 3d ed., Sambrook and Russell, CSHL Press (2001 ). Examples of methods may comprise but are not limited to, the CaCl2 chemical method, electroporation and the like. In particular, the constructs according to the present disclosuremay be delivered into the cells in vivo. Suitable methods of delivery of polynucleotide constructs are known in the art, and may comprise but are not limited to, viral vectors, nanoparticles, gold particles, lipoplexes and/or polyplexes.
[0092] The present disclosureprovides at least one method of preparing at least one HBV epitope-reactive exogenous TCR comprising: 1. (a) isolating at least one HBV-epitope reactive T cell from at least one HBV-exposed individual that resolved HBV infection; 2. (b) cloning at least one polynucleotide sequence encoding at least one a- and/or β-chain of at least one TCR from the HBV-epitope reactive T cell of step (a); 3. (c) delivering the polynucleotide sequence of step (b) to at least one cell; and 4. (d) incubating the cell under conditions suitable for expression of the HBV epitope-reactive exogenous T cell receptor by the cell.
[0093] In particular, the HBV-epitope reactive T cell of step (a) mentioned above may be HBc18-27 epitope reactive and/or HBs370-79 reactive.
[0094] According to one aspect, the present disclosureprovides at least one cell, at least one vector and/or at least one polypeptide according to all aspects of the present disclosure and invention, for use in the treatment of HBV infection and/or HBV-related hepatocellular carcinoma.
[0095] The present disclosurealso provides at least one method of treating HBV and/or inhibiting reactivation of HBV in at least one subject comprising administering to the subject at least one immunotherapeutically effective amount of cells, at least one vector and/or at least one polypeptide according to any aspect of the present disclosure and invention.
[0096] A further possible application of the HBV-TCR redirected approach is in patients with hepatocellular carcinoma (HCC). Chronic infection with HBV is the most important risk factor for the development of this liver tumour and integration of HBV-DNA into hepatocytes frequently occurs in tumour transformed cells. The therapy of HCC by conventional chemotherapy, radiation or surgical resection presents severe limitations. HBV-specific T cells have the potential to recognize and kill HCC cells expressing HBV antigens and adoptive transfer of HBV-TCR-redirected T cells may have the potential to obtain regression of HCC. Such results were obtained in nasopharyngeal carcinoma, a tumour expressing an Epstein-Barr Virus (EBV) protein, where infusion of EBV-specific CD8+ cells showed promising results. Vectors comprising polynucleotides encoding TCRs, and/or cells comprising at least one HBV-epitope reactive TCR may thus be useful in the treatment of HCC.
[0097] The present disclosure further provides at least one method of treating HBV-related hepatocellular carcinoma in at least one subject comprising administering to the subject at least one immunotherapeutically effective amount of cells, at least one vector and/or at least one polypeptide according to any aspect of the present disclosure and invention.
[0098] Vectors comprising polynucleotides encoding TCRs, and/or cells comprising at least one HBV-epitope reactive TCR prepared as described above, may be suitably administered to a subject to treat acute or chronic HBV infection or conditions (including, e.g., hepatocellular carcinoma) in the subject.
[0099] The cells expressing HBV-epitope reactive TCRs, vectors comprising polynucleotides encoding HBV-epitope reactive TCR and/or the polypeptide encoding the HBV-epitope reactive TCR may be prophylactically administered to at least one subject to inhibit reactivation of HBV infection. In particular, vectors of the disclosure may be administered to cells from at least one subject ex vivo. More in particular, modes of administration of polynucleotide and/or viral vectors will be those that specifically and/or predominantly deliver the TCR coding sequences to T cells and/or hematopoietic stem cells. In the case of a retroviral vector, it may be anticipated that suitable dosages will range from about 0.1 pg/108 cells to about 10 pg/108 cells, such as in the range from about 1 pg/108 cells to about 5 pg/108 cells. More in particular, such dosages may prevent or reduce HBV-related symptoms at least 50% compared to pre-treatment symptoms or compared to a suitable control. Treatment with at least one retroviral vector according to the present disclosure may palliate or alleviate HBV infection and/or at least one associated condition, and/or may reduce incidence of progression to chronic HBV-associated conditions, without providing a cure. In particular, treatment may be used to cure or prevent an acute or chronic HBV infection or an associated condition, including hepatocellular carcinoma.
[0100] In particular, the quantity of cells that make up the immunotherapeutically effective amount of cells to be administered depends on the subject to be treated. This may be dependent on but not limited to, the capacity of the individual's immune system to mount TCR-mediated immune response, the age, sexand weight of the patient and the severity of the condition being treated. The number of variables in regard to at least one individual's prophylactic or treatment regimen may be large, and a considerable range of doses may be expected. In particular, cells may be administered in at least one amount from 5 x 105 cells/kg body weight to 1 x 1010 cells/kg body weight. More in particular, 5 x 106 cells/kg body weight to 1 x 108 cells/kg body weight may be administered. The maximal dosage of cells and/or viral vector to be administered to the subject may be the highest dosage that does not cause undesirable and/or intolerable side effects. Suitable regimens for initial administration and additional treatments may also be contemplated and may be determined according to conventional protocols.
[0101] The present disclosure provides at least one use of at least one cell, at least one vector according and/or at least one polypeptide according to any aspect of the present invention and disclosure in the preparation of a medicament for treating HBV infection and/or HBV-related hepatocellular carcinoma.
[0102] Suitable solid or liquid medicament preparation forms may be, for example, granules, powders, tablets, coated tablets, (micro) capsules, suppositories, syrups, emulsions, suspensions, creams, aerosols, drops or injectable solutions in ampule form and also preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavourings, sweeteners or solubilizers are customarily used as described above. The medicaments may be suitable for use in a variety of drug delivery systems.
[0103] According to one aspect, the present invention provides at least one in vitro method for diagnosing at least one subject that is able to resolve HBV infection, the method comprising: 1. (a) providing at least one sample from at least one subject; 2. (b) detecting the presence of at least one polynucleotide comprising, substantially consisting of or consisting of at least one nucleic acid sequence selected from SEQ ID NO: 4, SEQ ID NO: 8, SEQ ID NO: 16 and SEQ ID NO: 20, SEQ ID NO: 43, SEQ ID NO: 51, and/or a homologue; and/or 3. (c) detecting the presence of at least one polypeptide comprising, substantially consisting of or consisting of the amino acid sequence of SEQ ID NO: 12, SEQ ID NO: 24, SEQ ID NO: 47, SEQ ID NO: 55, and/or a homologue thereof; wherein the presence of the polynucleotide and/or the polypeptide is indicative of the subject being able to resolve the HBV infection.
[0104] According to yet another aspect, the present invention provides at least one in vitro method for diagnosing at least one subject as having or as being at risk of having HBV infections and/or HBV related hepatocellular carcinoma, the method comprising: 1. (a) providing at least one sample from at least one subject; 2. (b) detecting the presence of at least one polynucleotide comprising, substantially consisting of or consisting of at least one nucleic acid sequence selected from SEQ ID NO: 4, SEQ ID NO: 8, SEQ ID NO: 16 and SEQ ID NO: 20, SEQ ID NO: 43, SEQ ID NO: 51, and/or a homologue thereof; and/or 3. (c) detecting the presence of at least one polypeptide comprising, substantially consisting of or consisting of the amino acid sequence of SEQ ID NO: 12, SEQ ID NO: 24, SEQ ID NO: 47, SEQ ID NO: 55, and/or a homologue thereof; wherein the absence of the polynucleotide and/or the polypeptide correlates with the likelihood of the subject as having or as being at risk for having HBV-infection and/or HBV-related hepatocellular carcinoma.
[0105] In particular, the test sample may be urine, blood, serum, sweat, and/or oral fluid samples. More in particular, the presence of at least one polynucleotide and/or polypeptide of the present invention may be determined using several methods known in the art. Some of the methods may comprise, but are not limited to, real time PCR, flow cytometry, PCR and/or geometric mean fluorescence intensity (MFI).
[0106] The present disclosure also provides at least one kit for detecting and/or treatment of HBV infection and/or HBV-related hepatocellular carcinoma, the kit comprising at least one cell, at least one vector and/or at least one polypeptide according to any aspect of the disclosure. In particular, the kit may comprise at least one manual providing instruction on howto use the kit.
Table 1. Sequences of HBV eptiope reactive exogenous TCR alpha 3 and beta 8 chains and naturally occurring HBc 18-27 epitope mutant peptides.
[0107] Having now generally described the invention, the same will be more readily understood through reference to the following examples which are provided by way of illustration, and are not intended to be limiting of the present invention.
EXAMPLES
[0108] Standard molecular biology techniques known in the art and not specifically described were generally followed as described in Sambrook and Russel, Molecular Cloning: A Laboratory Manual, Cold Springs Harbor Laboratory, New York (2001).
[0109] The following examples are provided to assist in a further understanding of the invention. The particular materials and conditions employed are intended to be further illustrative of the invention and are not limiting upon the reasonable scope of the appended claims.
Example 1
Cloning Hepatitis B virus specific T cell specific for HBV core 18-27 epitope (HBc18-27) [0110] Peripheral blood lymphocytes (PBL) were isolated from fresh blood obtained from a HBV patient who was able to resolve the disease and who was of HLAhaplotype HLA-A0201 herein referred to as "a resolved HLA-A201 HBV patient". The blood was first heparinised and PBL were isolated using Ficoll-Paque density gradient centrifugation. PBL were then washed three times in Hanks Balanced Salt Solution (HBSS) (Invitrogen, Carlsbad, CA), resuspended in Aim-V medium, 2% human AB serum (Invitrogen, Carlsbad, CA), stimulated with 1 μΜ of HBV core 18-27 epitope ("HBc18-27 peptide"; FLPSDFFPSV; SEQ ID NO: 25; Primm SRL, Milano, Italy) plus 20 U/ml interleukin-2- (R&amp;D systems, Minneapolis, MN) and plated in a 24-well plate at 4 x 106 cells/well for 10 days.
[0111] The HBc18-27-specific CD8+ T cells prepared as above were then labelled with phycoerythrin (PE)-conjugated HLA-A2 class I pentamers herein referred to as "HBc18-27-HLA-A201 pentamers" and/or "HLA-A2 pentamer" (Proimmune, Oxford, United Kingdom) bearing the HBc18-27 epitope for 30 min at 37°C and purified via magnetic cell sorting using anti-PE microbeads (Miltenyi Biotech, Surrey, United Kingdom). Following isolation, cells were labelled with Cy-chrome-conjugated anti-CD8+ (BD Pharmingen, San Diego, CA) and the percentage of stained T cells were determined by Fluorescence-activated cell sorting (FACS) analysis using a FACScan flow cytometer (BD Biosciences, SanDiego, CA) and analyzed using CellQuest software (BD Biosciences). The flow cytometry method used is further described in Gehring et al. The results are shown in Figure 2A.
[0112] The CD8+ and HBc18-HLA-A201 pentamer positive T cells as mentioned above, were then cloned using the limiting dilution assay as described in "Current Protocols in Immunology" Copyright © 2007 by John Wiley and Sons, Inc., and expanded in Aim-V 2% AB serum (Invitrogen, Carlsbad, CA), 20 U/ml IL-2, 10 ng/ml IL-7, and 10 ng/ml IL-15 (R&amp;D systems, Minneapolis, MN) with 1.5 pg/ml phytohemagglutinin (Sigma-Aldrich, Dorset, United Kingdom) using allogeneic irradiated PBL as feeder cells. Cells were plated at 1 cell/well on 96 well plates and wells positive for T cell growth were tested for HBc18-27 reactivity by intracellular cytokine staining for IFN-γ according to the protocol provided in Gehring et al.
[0113] Highly positive wells for peptide reactivity were then screened using the specific HBc18-27 HLA-A201 pentamer (Proimmune, Oxford, United Kingdom) and a T cell clone, referred to herein as C183 was selected for further study. The results, depicted in Figure 2A, demonstrate that 100% of CD8+ and HBc18-27-HLA-A201 pentamer- reactive T cells were detected. Clonality was tested using a panel of T cell receptor (TCR) Vp monoclonal antibodies (Beckman Coulter, Fullerton, CA). This procedure involved the use of a panel of Vp antibodies that stained all known Vp chain family members and where positive staining for only one Vp chain meant that it was highly likely that all of the T cells of that clone expressed the same TCR. Accordingly, the resultant TCR Vp distribution for C183, shown in Figure 2B demonstrates that all the T cells from C183 were Vp8 positive suggesting homogeneity (likely that they were all derived from a single cell) and thus clonality.
[0114] C183 thus was shown to be a clonal, CD8+, HBc18-27 specific cytotoxic T cell clone and was grown and maintained in Aim-V, 2% AB serum, 20 U/ml IL-2, 10 ng/ml IL-7, and 10 ng/ml IL-15 (R&amp;D Systems, Abingdon, United Kingdom) in a 5% CO 2 humidified incubator at 37° C.
Example 2
Recognition of tumor cells and primary human hepatocytes by C183 T cell clone [0115] HepG2 is a HLA-A2+, hepatocyte-like tumor cell line isolated from a primary hepatocellular carcinoma tumor (HCC). HepG2 cells (American Type Culture Collection, Rockford, MD) were grown and maintained in RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum (FBS), 20 mM HEPES, 0.5 mM sodium pyruvate, 100 U/ml penicillin, 100 pg/ml streptomycin, MeM amino acids, L-glutamine, MeM nonessential amino acids (Invitrogen, Carlsbad, CA), and 5 pg/ml Plasmocin (InvivoGen, San Diego, CA) to prevent mycoplasma contamination. Aderivative of this line, HepG2.105 (Kindly provided by Michael Nassal at the University of Freiberg) was engineered to express the entire HBV genome under the control of a doxycycline dependent regulator, which permits viral antigen to be expressed at various levels as described in Sun and Nassal, 2006. A vector control parental cell line, HepG2TA2-7 (Kindly provided by Michael Nassal at the University of Freiberg) was stably transfected with the doxycycline dependent regulator but not the HBV genome. Both cell lines HepG2.105 and HepG2TA2-7, were grown in Dulbecco's modified Eagle's medium supplemented with 10% tetracycline-approved FBS (BD Biosciences, San Diego, CA), 20 mM HEPES, 0.5 mM sodium pyruvate, 100 U/ml penicillin, 100 pg/ml streptomycin, MeM nonessential amino acids (Invitrogen), 200 pg/ml G418 sulfate, 80 pg/ml Plygromycin B (AutogenBioclear, Wiltshire, United Kingdom). Doxycycline (0.1 to 100 ng/ml, BD Biosciences) was added to cultures to regulate FIBV expression.
[0116] The standard HepG2 cells were labeled with 0.75 pM carboxyfluorescein succinimidyl ester (CFSE; Invitrogen) for 10 min at 37°C. Cells were washed, and CFSE-labelled targets were pulsed with 10'8 M FIBc18-27 peptide (Primm SRL, Milano, Italy) for 1 h on ice. Unlabelled (0 M HBc18-27 peptide) and CFSE-labelled (plus HBc18-27 peptide) HepG2 cells were mixed together in a 1:1 ratio, 2 x 105 cells of each PlepG2 cell line, in 24-well plates and incubated overnight to permit HepG2 adherence. To determine if C183 was capable of killing HCC tumor cells, C183 clones obtained from Example 1 were co-cultured with the HepG2 cells that were adhered to the wells ("targets"). A total of 4 x 105 of C183 clones were added to the targets and incubated for 5 h at 37°C. The resultant effector (C183):target (unlabelled + CFSE-labelled HepG2 cells) ratio was 1:1. After 5 h co-incubation, HepG2 cells were collected via trypsinization and cytotoxicity was determined by comparing ratio of peptide pulsed targets to unpulsed targets in wells +/- the addition of C183. The results shown in Figure 3 depict that CFSE negative cells (no HBc18-27 peptide) which were co-cultured with C183 remained and nearly all HepG2 cells which were PIBc18-27 and CFSE positive were eliminated by C183 as shown in the last column. The results indicate that the PIBc18-27 specific cytotoxic T cell clone, C183 was capable of killing tumor cells presenting HBV peptides.
[0117] Further, to determine if C183 can recognize endogenously processed antigen presented by HCC cells, C183 was co-cultured with HepG2.105 or HepG2TA2-7 control line and then tested for cytokine production and cytotoxic degranulation (CD107a+ staining), an indirect measure of killing. 1 x 105 cells of each HepG2.105 and HepG2TA2-7 cell line were seeded separately into 96-well plates and incubated overnight to permit HepG2 adherence. For cytotoxic degranulation assays, CD107a- PE antibody (BD Pharmingen, San Diego) was first added to all wells before a total of 1 x 105 of C183 clones were added to each well with the adhered HepG2.105 or HepG2TA2-7 control line and incubated for 5 h at 37°C. The resultant effector (C183):target (HepG2.105 or HepG2TA2-7 control line) ratio was 1:1. Following the incubation, C183 T cells were harvested, washed and stained with Cy-chrome conjugated anti-CD8+ (BD Pharmingen, San Diego, CA) then permeabilized and fixed using Cytofix/Cytoperm (BD Pharmingen, San Diego) according to the manufacturer's instructions. Cells were washed and incubated with PE-conjugated anti-human IFN-γ antibody (R&amp;D systems) for 30 min on ice then washed and analyzed by flow cytometry using the FACScan flow cytometer (BD Biosciences, SanDiego, CA) and analyzed using CellQuest software (BD Biosciences). The flow cytometry method used is further described in Gehring et al.
[0118] As shown in Figure 4A, C183 did not respond to the control cell line HepG2TA2-7 that was HBV negative. However, as shown in Figure 4B, co-culture of C183 with HepG2.105 cells expressing HBV, stimulated IFN-γ production and degranulation, indicating that C183 clone recognized the HBc18-27 epitope endogenously processed and presented by hepatocellular carcinoma cells.
[0119] In order to determine if C183 T cell clone can respond to naturally infected hepatocytes, primary hepatocytes were isolated from HLA-A2+ or HLA-A2-explanted chronic HBV livers and co-cultured with C183 according to the procedure described below.
[0120] Liver tissue was obtained from both HLA-A2+ or HLA-A2- explanted chronic HBV patients under informed consent according to ethical and moral guidelines of the institution. Tissue sections were collected in William's E medium (Sigma-Aldrich, Dorset, United Kingdom) and maintained at 4°C for a maximum of 2 h before cell isolation. Hepatocytes were isolated by enzyme perfusion described for use with human liver by Strain et al. (Strain, Ismail et al. 1991) with some modifications. Briefly, a section of liver of about 100 to 200 g was cut, and exposed vessels on a single surface were cannulated with 3-mm internal diameter tubing. The tissue was perfused sequentially at 50 ml/min with 500 ml PBS-HEPES wash solution to remove William's E medium, 500 ml PBS-HEPES-0.5 mM EGTA, and again with 500 ml PBS-HEPES wash solution (The solutions were obtained from Invitrogen Ltd.). Finally, 300 ml enzyme solution (0.05% collagenase (Roche, Indianapolis, IN), 0.012% hyaluronidase (Sigma, Dorset, United Kingdom), 0.025% Dispase II (Roche), 0.005% DNase (Roche) containing 5 mM CaCl2), maintained at 41 °C, was perfused with recirculation, and enzymatic digestion continued for 10 to 20 min until the liver was judged to be substantially softened. Tissue was mechanically dissociated in 200 ml Hanks' balanced salt solution (Invitrogen Ltd.) containing 10% FBS, 5 mM CaCl2· The cell suspension was filtered through a 60-pm cell strainer and pelleted at 37x g for 10 min at 4°C. Cells were washed three times in Hanks' balanced salt solution, after which viability and yield were assessed. Purified hepatocytes were used immediately after isolation. 100,000 primary hepatocytes/well were added to 96 well plates and 75,000 C183 clones were added to respective wells for 5 h with anti-CD107a-PE antibody (BD Pharmingen, San Diego) for degranulation plus 10 pg/ml brefeldin A (Sigma-Aldrich). Following the incubation, T cells were harvested, washed and stained with Cy-chrome conjugated anti-CD8+ (BD Pharmingen, San Diego, CA) then permeabilized and fixed using Cytofix/Cytoperm (BD Pharmingen, San Diego) according to the manufacturer's instructions. Cells were washed and incubated with FITC-conjugated anti-human IFN-γ antibody (R&amp;D systems, Abingdon, UK) for 30 min on ice then washed and analyzed by flow cytometry using the FACScan flow cytometer (BD Biosciences, SanDiego, CA) and analyzed using CellQuest software (BD Biosciences). The flow cytometry method used is further described in Gehring ef a/.
[0121] The results in Figure 5 show that C183 discharged cytotoxic granules, measured by CD107a staining, in response to HLA-A2 positive HBV infected primary hepatocytes but not to HLA-A2 negative HBV infected hepatocytes or HLA-A2+ uninfected hepatocytes, indicating that the C183 can recognize naturally infected primary human hepatocytes.
Example 3
Recognition of additional HLA-A2 subtypes and variant peptides [0122] Chronic HBV infection is predominantly an Asian problem and it is thus important to know if the HBc18-27 specific TCR can recognize HLA-A2 subtypes that dominate in the Asian population. HBV has a total of five genotypes (A, B, C, D and G). The HBc18-27 epitope expressed by HBV genotypes B and C (FLPSDFFPSI; SEQ ID NO:26) are the most prevalent in Asia. Genotypes A and D, most prevalent in Europe and United states, (FLPSDFFPSV; SEQ ID NO:25) are characterized by a valine at position 27 whereas genotypes B and C are characterized by an isoleucine at position 27. This can have a profound impact on the ability of HBV patients to present the HBc18-27 epitope or it can affect TCR recognition of the epitope.
[0123] The ability of C183 to recognize HBc18-27 epitope from genotypes B and C, and A and D presented by different subtypes of the HLA-A2 MHC class I family was determined using Epstein-Barr virus (EBV) transformed B cells with known subtypes of HLA-A2. (Kindly provided by Professor Chan Soh Ha at WHO Immunology and Training Research Centre, Singapore). EBV transformed B cells from any other source may be used in this procedure. The EBV transformed B cells (105 cells/well) were loaded with increasing concentrations (1 pM - 1000 pM) of HBc18-27 peptide from either HBV genotypes A/D or HBV genotypes B/C for 1 h at 25°C (HBV Genotype A/D = FLPSDFFPSV; Primm SRL; HBV genotype B/C = FLPSDFFPSI, Genscript, Piscataway, NJ) EBV B cells were washed with HBSS to remove excess peptide and co-cultured with 7.5x1C183 for 5 h in the presence of 10 ug/ml brefeldin A. IFN-γ production was measured by intracellular cytokine staining to determine T cell activation as described in Gehring et al.
[0124] The results are shown in Figure 6. The results show the ability of C183 to recognize the HBc18-27 epitope from four different HBV genotypes (A, B, C and D) and that the HBc18-27 epitopes were presented by the three most dominant HLA-A2 subtypes (HLA-A201, HLA-A206, and HLA-A207). The C183 T cell response is plotted at % IFN-y+ cells. The black bars represent the freguency of IFIM-y+ C183 T cells able to respond to the HBc18-27 epitope from HBV genotypes A/D (C18-V) whereas the grey bars represent the freguency of C183 T cells able to respond to HBc18-27 epitope from HBV genotypes B/C (C18-I) The data shows that HBc18-27 epitopes presented by (A) HLA-A201; (B) HLA-A206; and (C) HLA-A207 MHC class I molecules were recognized by C183. C183 recognized the HBc18-27 epitope with high sensitivity (< 1 pM) from all genotypes and the epitope was recognized almost egually when presented by the three most dominant HLA-A2 subtypes (A) A0201, (B) A0206 and (C) A0207.
[0125] To further show that the HBc-18-27 TCR transduced T cells recognize the HBc18-27 eptiope presented by multiple HLA-A2 subtypes and various mutants of the HBc18-27 epitope, EBV transformed B cells expressing HLA-A0201, - A0206 or -A0207 subtypes were loaded with increasing concentrations of HBV genotype AID HBc18-27 epitope (Figure 12 (A)) and HBV genotype B/C HBc18-27 epitope, (Figure 12 (B)) (sequence provided in Table 1) and co-cultured with HBc-18-27 TCR transduced T cells for 5h. T cell activation was measured by intracellular cytokine staining for IFN-γ. The results as shown in Figures 12 (A) and (B) show that the HBc-18-27 TCR transduced T cells recognize the HBc18-27 eptiope presented by multiple HLA-A2 subtypes.
[0126] Figure 12 (C) shows the results when HLA-A2+ T2 cells were loaded with each of the mutant peptides of HBc18-27 epitope and co-cultured with HBc-18-27 TCR transduced T cells for 5 h. T cell recognition of the mutant peptides was determined by intracellular cytokine staining for IFN-y. The sequences of the mutant peptides are provided in Table 1. As can be seen from the results, the HBc-18-27 TCR transduced T cells recognized various mutants of the HBc18-27 epitope. The Unstim 1 and
Unstim 2 refer to the control where the HLA-A2+ T2 cells were not loaded with any peptide.
Example 4
Isolation of C183 T cell receptor alpha and beta chain DNA
[0127] Total RNA was isolated from 5x10® C183 clones using TRIzqI (Invitrogen). TCR alpha and beta chain cloning was performed on a contract basis by Primm SRL (Milano, Italy) and supplied in Topo blunt II cloning vector for downstream cloning.
[0128] The sequence analysis revealed the presence of one TCR beta chain, Ν/β8.2, and one TCR alpha chain, Va3 in C183. Further DNA sequence analysis using the Immunogenetics V-Quest algorithm (http://imgt.cines.fr/IMGT_vquest/share/textes/) identified 3 unique Complementarity Determining Region (CDR) in the Va3 chain (CDR1a: SEQ ID NO:9, CDR2a: SEQ ID NO:10, CDR3a: SEQ ID NO: 11) and the three unique CDR in the Vp8.2 chain (CDR1p: SEQ ID NO:21, CDR2p: SEQ ID NO:22, CDR3p: SEQ ID NO:23).
Example 5
Retroviral constructs containing hBc18-27 specific TCR (HBc18-27 TCR) and lymphocyte transduction [0129] TCR Va3 and TCR \/β8.2 cDNA prepared according to Example 4 above, were cloned individually into retroviral vector MP71 as described in Engels, Cam et al. 2003. (Kindly provided by Professor Hans Stauss, Royal Free and UCL Medical School, London, UK) using a 5' Not-1 site and 3' BsrG1 sites (New England Biolabs, Ipswich, MA). TCR Va3, νβ8.2 and MP71 vector were digested for 1 h at 37°C with 10 U of each enzyme. Digested products were isolated on 1 % agarose gel by electrophoresis and purified using Qiagen gel extraction kit. TCR chains and MP71 vector were mixed in 1:1 ratio and ligated overnight at 4°C with T4 DNA ligase (Promega) to form retroviral constructs-MP71-TCR Va3 and MP71-TCR Vp8.2. These constructs were then sequenced to confirm DNA insert was correct.
[0130] To determine the expression and function of the cloned TCR, the constructs were used for preparation of retroviral supernatant which was further used for T cell transduction as explained below. Expression of inserted alpha and beta chains was driven by viral long terminal repeats (LTR) found in each construct. Retroviral supernatants were prepared using a transient transfection method which comprised using Phoenix amphotropic packaging cell line (Clontech Laboratories, US) that was first plated at 2x10® cells/dish and allowed to adhere for 24 h in Iscove's Modified Dulbecco's Medium (IMDM), 10% FBS, 25 mM HEPES, Glutamax (Invitrogen) and 5 ug/ml Plasmocin (Invivogen). Cells were transiently co-transfected using the CaCl2 method for 24 h with 9 pg each of MP71-TCR Va3, MP71-TCR Ν/β8.2 together with 6 pg of amphotropic envelope (kindly provided by Professor Hans Stauss, Royal Free and UCL Medical School, London, UK) IMDM was then replaced with Aim-V 2% human AB serum and phoenix cells were incubated for an additional 24 h before retroviral supernatants were collected for transduction.
[0131] Healthy donor and chronic HBV patient peripheral blood lymphocytes (PBL) were used to test expression and functionality of the cloned HBc18-27 TCR. Peripheral blood mononuclear cells (PBMC) were isolated from volunteers under informed consent by ficoll density gradient centrifugation. PBMC were stimulated with 600 U/ml interleukin-2 (IL-2; R&amp;D Systems) and 50 ng/ml anti-CD3 (OKT-3; eBioscience, San Diego, CA) for 48 h. Untreated 24 well tissue culture plates were coated with 30 ug/ml Retronectin (Takara Bio, Otsu Shiga, Japan) overnight at 4°C. Wells were then washed with HBSS and blocked with PBS 2% BSAfor 30 min.
Lymphocytes were harvested, washed, counted and 5x10® cells plated into retronectin coated wells and mixed with the retroviral supernatants collected as described above. Mock transduced cells which were included as negative control, were cultured with the supernatant from Phoenix cells that were not transfected with retroviral vectors. IL-2 was added to wells to final concentration of 600 U/ml. The lymphocytes were incubated for 24 h in the retroviral supernatant, after which time the medium was replaced with Aim-V 2% human AB serum plus 100 U/ml IL-2. Transduced and mock transduced cells were grown for additional 3 days. After 3 days, the cell surface expression of the TCR was measured in both the TCR transduced and mock transduced cells by immunofluorescence staining. CD8-PE-Cy7 (BD Biosciences), νβ8-ΡΕ expression (Beckman Coulter) and HLA-A201-HBc18-PE pentamer (Proimmune) staining were quantified by flow cytometry. Flow cytometry was performed on stained cells using a FACs Canto flow cytometer (BD Biosciences), and data were analyzed with the FACs Diva program (BD Biosciences).
[0132] As shown in Figure 7A, expression of νβ8 was significantly increased in HBc18-27 TCR transduced lymphocytes compared to mock transduced cells, which expressed only endogenous levels of Ν/β8. Ν/β8 expression was observed in both CD8+ and CD8- cells. To properly bind pentamer requires the cooperative interaction of a correctly paired, introduced alpha and beta chain as well as the co-receptor CD8. The CD8+ HBc18-27 TCR transduced T cells bound HBc18-27-HLA-A201 pentamers indicating that the Va3 TCR chain was also expressed and the introduced TCR was correctly paired on the cell surface of transduced lymphocytes (Figure 7B).
[0133] The HBc18-27 TCR transduction efficiency is confirmed in Figure 7 (C) which show the mean frequency of νβ8+ T cells from 5 healthy, 5 FIBeAg+ chronic FIBV patients (HBV DNA> 107 copies/ml), and 5 FIBeAg- chronic FIBV patients (HBV DNA< 106 copies/ml). Figure 7 (D) which shows the mean frequency of HBc18-27-A201 specific pentamer+ T cells from 5 healthy, 5 HBeAg+ chronic FIBV patients (FIBV DNA> 107 copies/ml), and 5 FIBeAg- chronic FIBV patients (FIBV DNA< 106 copies/ml) confirms that the transduced HBc18-27 TCR was correctly paired and bound HBc18-27-HLA-A201 pentamers. These data demonstrate that T cells from both healthy donors and chronic FIBV patients could be transduced with the FIBc18 TCR with similar efficiency.
Example 6
Function ofHBc18-27 TCR transduced T cells [0134] To confirm that the introduced FIBc18-27 TCR was functional, FILA-A2 positive T2 cells (American Type Culture Collection (Rockford, MD)) were used to stimulate FIBc18-27 TCR transduced or mock transduced lymphocytes. T2 cells were cultured in RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum (FBS), 20 mM FIEPES, 0.5 mM sodium pyruvate, 100 U/ml penicillin, 100 pg/ml streptomycin, MeM amino acids, Glutamax, MeM nonessential amino acids (Invitrogen Ltd) and 5 pg/ml Plasmocin (InvivoGen, San Diego, CA). The T2 cells were pulsed with 1 pg/ml FIBc18-27 peptide for 1 hr and then washed to remove excess peptide.
[0135] In order to stimulate the HBc18-27 TCR transduced lymphocytes, referred to herein as HBc18-27 TCR transduced T cells, that were prepared according to Example 5, the FIBc18-27 TCR transduced or mock transduced T cells were co-cultured overnight with the two different types of T2 cells (pulsed with HBc18-27 peptide and not pulsed) at 37°C with 2 ug/ml brefeldin A. The FIBc18-27 TCR transduced and mock transduced T cells were then washed and stained with anti-CD8-PE-Cy7 (BD Pharmingen, San Diego, CA) fixed and permeablized using cytofix/cytoperm (BD Biosciences) according to manufacturer's instructions. T cells were then stained for TNF-a-Alexa 488, IL-2-PE and IFN-y-APC (BD Biosciences) for 30 min on ice. Cells were washed and analyzed by flow cytometry using FACs Canto flow cytometer (BD Biosciences), and data were analyzed with the FACs Diva program (BD Biosciences).
[0136] The results are shown in Figure 8A. The mock transduced T cells failed to produce any cytokines (i.e. IFN-y, TNF-α or IL-2) in the presence or absence of HBc18-27 epitope. HBc18-27 TCR transduced T cells did not respond to T2 cells in the absence of peptide (Figure 8A, row 1) but produced IFN-y, TNF-α and IL-2 in response to HBc18-27 peptide loaded T2 cells (Figure 8A, row 3) indicating that the introduced HBc18-27 TCR was functional when expressed in primary T cells. Furthermore, the introduced HBc18-27 TCR was functional in CD8- cells (CD4+ cells) as well as CD8+, expanding its potential usefulness for immunotherapy because CD4+ T cells provide additional IL-2 to maintain T cell survival and co-stimulatory ligands such as CD40L, which has the potential to boost B cell responses.
[0137] Figure 8 (B) shows that FIBc18-27 TCR transduced T cells from chronic FIBV patients are equally functional to transduced T cells from healthy donors. Mean frequency of IFN-y, TNF-α and IL-2 positive cells, as seen in figure 8A, from 5 healthy, 5 FIBeAg+ chronic FIBV patients (FIBV DNA > 107 copies/ml), and 5 FIBeAg- chronic FIBV patients (FIBV DNA< 106 copies/ml) are shown. The results show that T cells transduced with FIBc18-27-A201 TCR become multifunctional with similar frequencies of cytokine positive cells in each patient group.
Example 7
Affinity of HBc18-27 TCR transduced T cells [0138] Due to the possible decreased level of expression of the introduced HBc18-27 TCR, it was possible that the engineered T cells may have a lower affinity for the actual viral epitope compared to the original T cell clone, C183. To test this hypothesis, HLA-A2 positive T2 cells (American Type Culture Collection, Rockford, MD) were pulsed with HBc18-27 peptide at concentrations ranging from 1 fg/ml to 1 pg/ml according to the protocol described in Example 6. Peptide loaded T2 cells were co-cultured with TCR transduced T cells from 5 healthy donors, 5 HBeAg+ chronic HBV patients (HBV DNA > 107 copies/ml), and 5 HBeAg- chronic HBV patients (HBV DNA< 106 copies/ml) and the original T cell clone C183 for 5 h. T cell activation was measured by intracellular cytokine staining for IFN-γ. Results are displayed at percent of maximum IFN-γ response because the frequency of HBc18-27 TCR transduced cells varied among patients while C183 was always 100% specific for the HBc18-27 epitope. The results showed that CD8+ HBc18-27 TCR transduced T cells displayed an affinity identical to C183 down to 1 pg/ml (Figure 9A).
[0139] The affinity of HBc18-27 TCR transduced CD4+ T cells was determined in the same assay. CD8- HBc18-27 specific cells were considered CD4+ T cells because they were capable of responding to the HBc18-27 epitope in an antigen specific, dose dependent fashion. CD4+ HBc18-27 TCR transduced T cells showed slightly lower affinity compared to C183. The difference in the affinity to the viral epitope between CD8+ and CD4+ T cells may likely be due to CD8+ co-receptor binding, which stabilizes TCR interaction with the HLA-A2 molecule. The CD8+ co-receptor is absent on CD4+ T cells and therefore affinity is slightly reduced but still highly sensitive.
Example 8
Codon optimization of HBc18-27 TCR DNA sequence [0140] Due to the presence of redundancy in the genetic code, most amino acids can be coded for by multiple codons, which are used with different frequencies and efficiencies within organisms and can actually result in less efficient translation of proteins. Due to genetic variation and the recombination events that occur during T cell development, DNA encoding for the HBc18-27 TCR may be comprised of codons that are not optimally translated within humans. Therefore, codon optimization for both the νβ8.2 and Va3 chains of the HBc18-TCR was done to result in a more efficient expression of HBc18-27 TCR and increased sensitivity and function (Service by Genscript, Piscataway, NJ).
[0141] Codon optimized Va3 and νβ8.2 were cloned individually into MP71 vector using 5'Not-1 and 3'BsrG1 restriction enzyme sites as described above. In order to confirm that the codon optimized TCR, referred to herein as HBc18-Opt-TCR, was still capable of being expressed, PBL were transduced with the HBc18-Opt-Va3 and HBc18-OptA^8.2 TCR constructs as described above. For the HBc18-27 wild type TCR, referred to herein as HBc18-WT-TCR, PBL were also transduced with the HBc18-WT-νβ8.2 (MP71-TCR νβ8.2 from example 7) and HBc18-WT-Va3 (MP71-TCR Va3 from example 7) for comparison. HBc18-27 transduced PBL were analyzed for νβ8 expression and the ability to bind HBc18-HLA-A201 pentamers (Proimmune, Oxford, United Kingdom) by flow cytometry 3 days after transduction. Results showed that the frequency of νβ8+ T cells was similar between PBL transduced with HBc18-Opt-TCR (21.5%) compared to HBc18-WT-TCR (21.1 %) transduced cells (Figures 10A) [0142] The amount of νβ8 expressed in the HBc18-Opt-TCR transduced lymphocytes and HBc18-WT-TCR transduced lymphocytes was measured by mean fluorescence intensity (MFI) using the FACs Diva software (BD Biosciences) (Figure 10B). Isotype values were subtracted to determine specific MFI. The results showed that the amount of νβ8 expressed was almost identical in both the HBc18-Opt-TCR transduced lymphocytes and HBc18-WT-TCR transduced lymphocytes (Figure 10B).
[0143] To determine the ability to bind HBc18-HLA-A201 pentamers, the HBc18-Opt-TCR transduced lymphocytes and HBc18-WT-TCR transduced lymphocytes were labelled with anti-CD8-PE-Cy7 (BD Pharmingen, San Diego, CA) and HBc18-HLA-A201 pentamers (Proimmune, Oxford, United Kingdom). The percentages of stained lymphocytes were determined by Fluorescence-activated cell sorting (FACS) analysis using a FACs Canto flow cytometer (BD Biosciences, SanDiego, CA) and analyzed using FACs Diva software (BD Biosciences). The results showed that the frequency of HBc18-HLA-A201 pentamer positive cells was two fold higher in cells transduced with HBc18-Opt-TCR than the HBc18-WT-TCR (Figure 10C), suggesting that the Va3 chain may be expressed more efficiently from the codon optimized construct compared to the wild type sequence. Because νβ8 staining was equivalent in Figure 10A increased pentamer binding is likely related to better expression of the Va3 chain to increase expression of correctly paired HBc18-27 TCR on the cell surface.
[0144] Following confirmation that the HBc18-Opt-TCR was properly expressed in the transduced T cells, we tested functionality of the HBc18-Opt-TCR compared to the HBc18-WT-TCR. HBc18-Opt-TCR and HBc18-WT-TCR or mock transduced T cells were stimulated overnight with HLA-A2 positive T2 cells in the presence or absence of HBc18-27 peptide plus 2 pg/ml brefeldin A according to the protocol described in Example 6. T cells were then labelled with anti-CD8 PE-Cy7 (BD Pharmingen, San Diego, CA) and fixed and permeablized using cytofix/cytoperm according to manufacturer's instructions (BD Biosciences). Cells were then stained with anti-IFN-y-APC (BD Biosciences) for 30 min on ice and T cell IFN-γ production was analyzed by flow cytometry.
[0145] The results show that there was nearly a two fold increase in the frequency of IFN-γ positive T cells transduced with the HBc18-Opt-TCR compared to T cells transduced with the HBc18-WT-TCR (Figure 11). Mock transduced cells were negative in the presence of absence of HBc18-27 peptide. Therefore, the increase in pentamer positive cells correlated with an increase in functional cells able to respond to peptide loaded targets.
Example 9
Abc18-27 TCR transduced T cells kill HCC cell lines which endogenously express HBV proteins or are loaded with the HBV18-27 peptide.
[0146] HBc18-27 TCR transduced T cells from each patient group (healthy, HBeAg(-), HBeAg(+)) were co-cultured with DiOC labeled HepG2 cells expressing the entire HBV genome (HBV-HepG2) or the parental control cell line (Ctrl-HepG2) at EffectonTarget ratio of 1:1 for 6 h in the presence of Propidium Iodide (PI). Dying targeT cells were DiOC+/PI+. T cells were excluded from the analysis by staining with anti-CD11a-APC (BD Bioscience). Results are representative of 3 experiments. Effector T cells were considered to be IFN-y+/CD8+ cells determined by intracellular cytokine staining after stimulation with peptide pulsed T2 cells. The results as shown in Figure 13 (A) show that HBc18-27 TCR transduced T cells from healthy donors or chronic HBV patients can kill HCC cell lines which endogenously express HBV proteins. This suggests that HBc18 TCR transduced T cells could recognize tumor cells from patients expressing the HBV core antigen.
[0147] To determine if TCR transduced T cells could kill multiple HLA-A2+ HCC cell lines, DiOC labeled HCC cell lines (HepG2 were obtained from ATCC; SNU-387, SNU-368 were obtained from the Korean cell line bank) were loaded with increasing concentrations of HBc18-27 peptides and co-cultured with HBc-18-27 TCR transduced T cells at Effector to target ratio of 1:1 for 6 h in the presence of Propidium Iodide (PI). Dying targeT cells were DiOC+/PI+. T cells were excluded from the analysis by staining with anti-CD11a-APC (BD Bioscience). Results are representative of 3 experiments. Effector T cells were considered to be IFN-y+/CD8+ cells determined by intracellular cytokine staining after stimulation with peptide pulsed T2 cells. The results as shown in Figure 13 (B) showed that HBc18-27 TCR transduced T cells are able to kill multiple HLA-A2+ HCC cell lines.
Example 10
Cloning Hepatitis B virus specific T cell specific for HBV envelope 370-79 eptitope (HBs370-79) [0148] Peripheral blood lymphocytes (PBL) were isolated and prepared according to the method of Example 1 and stimulated with 1 μΜ of HBV envelope 370-79 epitope ("HBs370-79 peptide"; SIVSPFIPLL; SEQ ID NO: 56; Primm SRL, Milano, Italy) plus 20 U/ml interleukin-2- (R&amp;D systems, Minneapolis, MN) and plated in a 24-well plate at 4 x 106 cells/well for 10 days.
[0149] The HBs370-79-specific CD8+ T cells were enriched using IFN-γ capture assay according to manufacturer's instructions. (Miltenyi Biotech, Surrey, United Kingdom).
[0150] The IFN-y+ HBs370-79- specific T cells were then cloned using the limiting dilution assay as described in "Current Protocols in Immunology" Copyright © 2007 by John Wiley and Sons, Inc., and expanded in Aim-V 2% AB serum (Invitrogen, Carlsbad, CA), 20 U/ml IL-2, 10 ng/ml IL-7, and 10 ng/ml IL-15 (R&amp;D systems, Minneapolis, MN) with 1.5 pg/ml phytohemagglutinin (Sigma-Adrich, Dorset, United Kingdom) using allogeneic irradiated PBL as feeder cells. Cells were plated at 1 cell/well on 96 well plates and wells positive for T cell growth were tested for HBs370-79 reactivity by intracellular cytokine staining for IFN-γ according to the protocol provided in Gehring et al. T cell clone, referred to herein as E10-1 D was selected for further study.
[0151] E10-1 D HBs370-79 specific cytotoxic T cell clone was grown and maintained in Aim-V, 2% AB serum, 20 U/ml IL-2, 10 ng/ml IL-7, and 10 ng/ml IL-15 (R&amp;D Systems, Abingdon, United Kingdom) in a 5% CO2 humidified incubator at 37° C.
Example 11
Affinity and Function of HBs370-79 specific TCR transduced T cells and Recognition of genotype variant peptides [0152] Experiments similar to that described in Example 6 were carried out with HBs370-79 specific TCR transduced T cells. The E10-1D T cell clone or HBs370-79 TCR transduced T cells, recognize HBs370-79 epitiope loaded HLA-A2+ T2 cells. T2 cells were loaded with increasing concentrations of HBs370-79 peptide and co-cultured with E10-1D or HBs370-79 TCR transduced T cells for 5 h and stained for IFN-γ. Data presented as percent maximum response to normalize differing frequency of each line. The results as shown in Figure 14 (A) show that FIBs370-79 specific TCR is functional but that FIBs370 TCR transduced T cells were lower affinity than the original T cell clone E10-1D.
[0153] Experiments similar to that described in Example 3 were also carried out with HBs370-79 specific TCR transduced T cells. FIBs370-79 TCR transduced T cells recognize genotypic variants of the HBs370-79 epitope (The sequences of the genotypic variants are provided in Table 2). FIBs370-79 TCR transduced T cells were co-cultured with FILA-A2+ T2 cells loaded with 1 pg/ml peptide of each genotype for 5 h and activation was assessed by CD107a labeling and staining for IFN-γ. Genotype A &amp; D sequences are identical. The results as shown in Figure 14 (B) show that FIBs370-79 specific TCR recognizes genotypic variants of the HBs370-79 epitope.
References: [0154]
Engels, B., H. Cam, et al. (2003). "Retroviral vectors for high-level transgene expression in T lymphocytes." Flum Gene Ther 14(12): 1155-68.
Gehring A.J et al. (2007). The Level of Viral Antigen Presented by Hepatocytes Influences CD8 T cell Function. J Virol.; 81(6): 2940-2949.
Karlin S. and Altschul S.F, (1990). Methods for assessing the statistical significance of molecular sequence features by using general scoring schemes. Proc Natl Acad Sci U S A.; 87(6): 2264-2268.
Maini, M. and Bertoletti A (2006) "The effect of pathogens on the immune system: Viral hepatitis". In immunodominance: The choice of the Immune System, J.A. Frelinger, ed (Weinheim: Wiley-VCH).
Molecular Cloning: A Laboratory Manual, 3d ed., Sambrookand Russell, CSFIL Press (2001).
Schumacher et. al., (2002), Nat Rev. Immunol, Vol 2: 512.
Strain, A. J., et al. 1991. Native and recombinant human hepatocyte growth factors are highly potent promoters of DNAsynthesis in both human and rat hepatocytes. J Clin Invest 87:1853-7.
Sun, D. and M. Nassal (2006). "Stable HepG2- and Huh7-based human hepatoma cell lines for efficient regulated expression of infectious hepatitis B virus." J Hepatol 45(5): 636-45.
SEQUENCE LISTING
[0155]
<110> A*STAR
Agency for Science Technology and Research <120> HBV Epitope-Reactive Exogenous T Cell Receptor and Uses Thereof <130> FP4671 <160> 58 <170> Patentln version 3.4 <210> 1 <211> 21
<212> DNA <213> Homo sapiens <220> <221 > misc_feature
<223> Complementarity Determining Region 1 (CDR1) for alpha 3 chain of vwld type TCR <400 1 aaaactagta taaacaattt a 21 <210> 2 <211> 24
<212> DNA <213> Homo sapiens <220> <221 > misc_feature
<223> Complementarity Determining Region 2 (CDR2) for alpha 3 chain of vwld type TCR <400> 2 ttaatacgtt caaatgaaag agag 24 <210> 3 <211> 42
<212> DNA <213> Homo sapiens <220> <221 > misc_feature
<223> Complementarity Determining Region 3 (CDR3) for alpha 3 chain of vuld type TCR <400>3 tgtgctacgt ggctctctgg ttctgcaagg caactgacct tt 42 <210> 4 <211> 822
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Alpha chain of wild type TCR <220> <221 > Misc_feature
<223> Alpha 3 chain of wild type TCR <400> 4
atggaaactc tcctgggagt gtctttggtg attctatggc ttcaactggc tagggtgaac SO agtcaacagg gagaagagga tcctcaggcc ttgagcatcc aggagggtga aaatgccacc 120 atgaactgca gttacaaaac tagtataaac aatttacagt ggtatagaca aaattcaggt 180 agaggccttg tccacctaat tttaatacgt tcaaatgaaa gagagaaaca cagtggaaga 240 ttaagagtca cgcttgacac ttccaagaaa agcagttcct tgttgatcac ggcttcccgg 300 gcagcagaca ctgcttctta cttctgtgct acgtggctct ctggttctgc aaggcaactg 360 acctttggat ctgggacaca attgactgtt ttacctgata tccagaaccc tgaccctgcc 420 gtgtaccagc tgagagactc taaatccagt gacaagtctg tctgcctatt caccgatttt 480 gattctcaaa caaatgtgtc acaaagtaag gattctgatg tgtatatcac agacaaaact 540 gtgctagaca tgaggtctat ggacttcaag agcaacagtg ctgtggcctg gagcaacaaa 600 tctgactttg catgtgcaaa cgccttcaac aacagcatta ttccagaaga caccttcttc 660 cccagcccag aaagttcctg tgatgtcaag ctggtcgaga aaagctttga aacagatacg 720 aacctaaact ttcaaaacct gtcagtgatt gggttccgaa tcctcctcct gaaagtggcc 780 gggtttaatc tgctcafcgac gctgcggctg tggtccagct ga 822 <210> 5 <211> 21 <212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 1 (CDR1) for alpha 3 chain of optimised TCR <400>5 aagacatcaa tcaacaactt g 21 <210> 6 <211> 24 <212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 2 (CDR2) for alpha 3 chain of optimised TCR <400>6 ctgattcgga gtaatgagcg ggaa 24 <210> 7 <211> 42 <212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 3 (CDR3) for alpha 3 chain of optimised TCR <400>7 tgtgctacat ggcfcgagtgg cagcgcacgg caattgactt tt 42 <210> 8 <211 > 822
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Alpha 3 chain of optimised TCR <400>8 atggagaccc ttctgggagt gtcccfccgtg attctgtggc tgcagcttgc tcgggtgaat 60 t.ctcagcagg gcgaggaaga cccgcaggcc cttagcattc aggaagggga gaacgctacc 120 atgaattgct catacaagac atcaatcaac aacttgcagt ggtaccgtca gaactctggg 180 agaggactcg tgcacctgat cctgattcgg agtaatgagc gggaaaaaca ctctggaagg 240 ctgagggtga ccctcgatac ctctaaaaaa tcctcctccc tgctgataac cgccagcagg 300 gccgccgaca ccgcttccta cttctgtgct acatggctga gtggcagcgc acggcaattg 360 acttttggga gtggcactca gctgacagtg ctgcccgaca tccagaatcc agatcacgca 420 gtgtatcagc tgagagactc aaagtcaagt gacaagagtg tgtgcctgtt cactgatttt 480 gactctcaga ccaacgtctc tcagtctaag gacagcgacg tttacatcac tgacaaaact 540 gtgctggaca tgcgcagtat ggactttaaa tcaaattccg ccgtggcfctg gagcaataag 600 tctgacttcg cctgtgctaa tgcttttaat aactccatca ttccggagga tacatttttc 660 cctagccccg agtcatcctg cgacgtgaag ctggtggaga agtcattcga gaccgacacc 720 aatcttaact ttcagaacct gtccgttatc gggtttagaa tcctgctgct gaaggttgcc 780 ggattcaacc tgcttatgac gttgcgcctg tggtccagct ga 822 <210> 9 <211 > 7
<212> PRT <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 1 (CDR1) for alpha 3 chain of TCR <400>9
Lys Thr Ser lie Asn Asn Leu 1 5 <210> 10 <211 > 8 <212> PRT <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 2 (CDR2) for alpha 3 chain of TCR <400> 10
Leu Ile Arg Ser Asn Glu Arg Glu 1 5 <210> 11 <211> 14
<212> PRT <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 3 (CDR3) for alpha 3 chain of TCR <400> 11
Cys Ala Thr Trp Leu Ser Gly Ser Ala Arg Gin Leu Thr Phe 15 10 <210> 12 <211>273
<212> PRT <213> Homo sapiens <220>
<221 > Misc_feature <223> Alpha 3 chain of TCR <400> 12
Met Glu Thr Leu Leu Gly Val Ser Leu Val lie Leu Trp Leu Gin Leu 15 10 15
Ala Arg Val Asn Ser Gin. Gin Gly Glu Glu Asp Pro Gin Ala Leu Ser 20 25 30 lie Gin Glu Gly Glu Asn Ala Thr Met Asn Cys Ser Tyr Lys Thr Ser 35 40 45
lie Asn Asn Leu Gin Trp Tyr Arg Gin Asn Ser Gly Arg Gly Leu Val 50 55 SO
His Leu lie Leu lie Arg Ser Asn Glu Arg Glu Lys His Ser Gly Arg 65 70 75 80
Leu Arg Val Thr Leu Asp Thr Ser Lys Lys Ser Ser Ser Leu Leu lie 85 90 95
Thr Ala Ser Arg Ala Ala Asp Thr Ala Ser Tyr Phe Cys Ala Thr Trp 100 105 110
Leu Ser Gly Ser Ala Arg Gin Leu Thr Phe Gly Ser Gly Thr Gin Leu 115 120 125
Thr Val Leu Pro Asp lie Gin Asn Pro Asp Pro Ala Val Tyr Gin Leu 130 135 140
Arg Asp Ser Lys Ser Ser Asp Lys Ser Val Cys Leu Phe Thr Asp Phe 145 150 155 160
Asp Ser Gin Thr Asn Val Ser Gin Ser Lys Asp Ser Asp Val Tyr lie 165 170 175
Thr Asp Lys Thr Val Leu Asp Met Arg Ser Met Asp Phe Lys Ser Asn 180 185 190
Ser Ala Val Ala Trp Ser Asn Lys Ser Asp Phe Ala Cys Ala Asn Ala 195 200 205
Phe Asn Asn Ser lie lie Pro Glu Asp Thr Phe Phe Pro Ser Pro Glu 210 215 220
Ser Ser Cys Asp Val Lys Leu Val Glu Lys Ser Phe Glu Thr Asp Thr 225 230 235 240
Asn Leu Asn Phe Gin Asn Leu Ser Val lie Gly Phe Arg lie Leu Leu 245 250 255
Leu Lys Val Ala Gly Phe Asn Leu Leu Met Thr Leu Arg Leu Trp Ser 260 265 270
Ser <210> 13 <211> 21 <212> DNA <213> Homo sapiens <220> <221 > Misc_feature <223> Complementarity Determining Region 1 (CDR1) for beta 8 chain of wild type TCR <400> 13 atttcaggac acgactacot t 21
<210> 14 <211> 24 <212> DNA <213> Homo sapiens <220> <221 > Misc_feature <223> Complementarity Determining Region 2 (CDR2) for beta 8 chain of wild type TCR <400> 14 tactttaaca acaacgttcc gata 24 <210> 15 <211> 45 <212> DNA <213> Homo sapiens <220> <221 > Misc_feature <223> Complementarity Determining Region 3 (CDR3) for beta 8 chain of wild type TCR <400> 15 tgtgccagca gcaatcgggc gagctcctac aatgagcagt tcttc 45 <210> 16 <211> 942 <212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Beta 8 chain of wild type TCR <400> 16 atggactcct ggaccctctg ctgtgtgtcc ctttgcatcc tggtagcaaa gcacacagat 60 gctggagtta tccaatcacc ccggcacgag gtgacagaga tgggacaaga agtgactctg 120 agatgtaaac caatttcagg acacgactac cttttctggt acagacagac catgatgcgg 180 ggactggagt tgctcattta ctttaacaac aacgttccga tagatgattc agggatgccc 240 gaggatcgat tctcagctaa gatgcctaat gcatcattct ccactctgaa gatccagccc 300 tcagaaccca gggactcagc tgtgtacttc tgtgccagca gcaatcgggc gagctcctac 360 aatgagcagt tcttcgggcc agggacacgg ctcaccgtgc tagaggacct gaaaaacgtg 420 ttcccacccg aggtcgctgt gtttgagcca tcagaagcag agatctccca cacccaaaag 480 gccacactgg tgtgcctggc cacaggcttc taccccgacc acgtggagct gagctggtgg 540 gtgaatggga aggaggtgca cagtggggtc agcacagacc cgcagcccct caaggagcag 600 cccgccctca atgactccag atactgcctg agcagccgcc tgagggtctc ggccaccttc 660 tggcagaacc cccgcaacca cttccgctgt caagtccagt tctacgggct ctcggagaat 720 gacgagtgga cccaggatag ggccaaacct gtcacccaga tcgtcagcgc cgaggcctgg 780 ggtagagcag actgtggctt cacctccgag tcttaccagc aaggggtcc~ gtctgccacc 840 atcctctatg agatcttgct agggaaggcc accttgtatg ccgtgctggt cagtgccctc 900 gtgctgatgg ccatggtcaa gagaaaggat tccagaggct ag 942 <210> 17 <211> 21
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature <223> Complementarity Determining Regionl (CDR1) for beta 8 chain of optimised TCR <400> 17 atctctgggc acgactacct g 21 <210> 18 <211> 24
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature <223> Complementarity Determining Region 2 (CDR2) for beta 8 chain of optimised TCR <400> 18 tattttaata acaatgtgcc tato 24 <210> 19 <211 >45 <212> DNA <213> Homo sapiens <220> <221 > Misc_feature <223> Complementarity Determining Region 3 (CDR3) for beta 8 chain of optimised TCR <400> 19 tgtgcctcct ccaaccgggc ctcctcttat aacgagcagt tcttc 45 <210>20 <211> 942 <212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Beta 8 chain of optimised TCR <400> 20 atggacagct ggacactgtg ctgcgtgagc ctgtgcattc tggtggccaa gcacaccgac 60 gccggcgtga tccagagccc tcgccacgag gtgaccgaaa tgggccagga ggtgacactg 120 cgctgcaagc caatctctgg gcacgactac ctgttctggt acaggcagac catgatgagg 180 ggcctggaac tgctgatcta ttttaataac aatgtgccta tcgatgactc tggcatgccc 240 gaggacaggt tctccgccaa gatgcccaac gccagcttct 'ccaccctgaa gatccagccc 300 tccgaaccta gggactccgc cgtgtacttc tgtgcctcct ccaaccgggc ctcctcttat 360 aacgagcagt tcttcggccc tggaacccgc ctgaccgtgc tggaggacc^ gaaaaatgtg 420 tttccccccg aggtggccgt gtttgaacca agcgaggccg agatcagcca cacacagaag 480 gccaccctgg tgtgtctggc caccggattc tatcccgatc acgtggagcc gagctggtgg 540 gtgaacggga aggaggtgca ctctggcgtg agcaccgacc ctcagccac- gaaagagcag 600 cccgccctga atgattctcg gtactgcctg tccagccgcc tgcgcgtgtc tgccaccttc 660 tggcagaacc ccagaaatca cttcaggtgc caggtgcagt tctatgggcr. gagcgagaac 720 gacgaatgga cccaggacag agccaagcct gtgacacaga tcgtgtctgc cgaagcctgg 780 ggcagagccg actgcggctt taccagcgag agctaccagc agggcgtgct gtccgccaca 840 attctgtacg agatcctgct gggaaaggcc acactgtacg ccgtgctggt gagcgccctg 900 gtgctgatgg ccatggtgaa gcggaaagac tcccggggct ga 942 <210>21 <211 > 7
<212> PRT <213> Homo sapiens <220 <221 > Misc_feature <223> Complementarity Determining Region 1 (CDR1) for beta 8 chain of TCR <400 21
Ile Ser Gly His Asp Tyr Leu 1 5 <210> 22 <211 > 8
<212> PRT <213> Homo sapiens <220> <221 > Misc_feature <223> Complementarity Determining Region 2 (CDR2) for beta 8 chain of TCR <400> 22
Tyr The Asn Asn Asn Val Pro Ile 1 5 <210> 23 <211 > 15
<212> PRT <213> Homo sapiens <220> <221 > Misc_feature <223> Complementarity Determining Region 3 (CDR3) for beta 8 chain of TCR <400> 23
Cys Ala Ser Ser Αεη Arg Ala Ser Ser Tyr Aan Glu Gin Phe Phe 15 10 15 <210> 24 <211 > 313
<212> PRT <213> Homo sapiens <220>
<221 > Misc_feature <223> Beta 8 chain of TCR <400> 24
Met Asp Ser Trp Thr Leu Cys Cys Val Ser Leu Cys Ile Leu Val Ala 15 10 15
Lys His Thr Asp Ala Gly Val Ile Gin Ser Pro Arg His Glu Val Thr 20 25 30
Glu Met Gly Gin Glu Val Thr Leu Arg Cys Lys Pro Ile Ser Gly His 35 40 45
Asp Tyr Leu Phe Trp Tyr Arg Gin Thr Met Met Arg Gly Leu Glu Leu 50 55 60
Leu Ile Tyr Phe Asn Asn Asn Val Pro Ile Asp Asp Ser Gly Met Pro 65 70 75 80
Glu Asp Arg Phe Ser Ala Lys Met Pro Asn Ala Ser Phe Ser Thr Leu 85 90 95
Lys Ile Gin Pro Ser Glu Pro Arg Asp Ser Ala Val Tyr Phe Cys Ala 100 105 110
Ser Ser Asn Arg Ala Ser Ser Tyr Asn Glu Gin Phe Phe Gly Pro Gly 115 120 125
Thr Arg Leu Thr val Leu Glu Asp Leu Lys Asn Val Phe Pro Pro Glu 130 135 140
Val Ala Val Phe Glu Pro Ser Glu Ala Glu Ile Ser His Thr Gin Lys 145 150 155 160
Ala Thr Leu Val cys Leu Ala Thr Gly Phe Tyr Pro Asp His Val Glu 165 170 175
Leu Ser Trp Trp Val Asn Gly Lys Glu Val His Ser Gly Val Ser Thr ISO 185 190
Asp Pro Gin Pro Leu Lys Glu Gin Pro Ala Leu Asn Asp Ser Arg Tyr 195 200 205
Cys Leu Ser Ser Arg Leu Arg Val Ser Ala Thr Phe Trp Gin Asn Pro 210 215 220
Arg Asn His Phe Arg Cys Gin Val Gin Phe Tyr Gly Leu Ser Glu Asn 225 230 235 240
Asp Glu Trp Thr Gin Asp Arg Ala Lys Pro Val Thr Gin Ile Val Ser 245 250 255
Ala Glu Ala Trp Gly Arg Ala Asp Cys Gly Phe Thr Ser Glu Ser Tyr 260 265 270
Gin Gin Gly Val Leu Ser Ala Thr Ile Leu Tyr Glu Ile Leu Leu Gly 275 280 285
Lys Ala Thr Leu Tyr Ala Val Leu Val Ser Ala Leu Val Leu Met Ala 290 295 300
Met Val Lys Arg Lys Asp Ser Arg Gly 305 310
<210> 25 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> HBc18-27 epitope (Genotype A/D/E/F); (WT C18-27) <400> 25
Phe Leu Pro Ser Asp Phe Phe Pro Ser Val 15 10
<210> 26 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> HBc18-27 epitope (Genotype B/C); 27I <400> 26
Phe Leu pro Ser Asp Phe Phe Pro Ser Ile 15 10
<210> 27 <211> 10 <212> PRT <213> Hepatitis B virus <220 <221 > Misc_feature <223> Natural Variant 1 of HBc18-27 epitope <400> 27 phe Leu Pro Asn Asp Phe phe Pro Ser val 15 10
<210> 28 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 2 of HBc18-27 epitope <400> 28
Phe Leu Pro Asn Asp Phe She Pro Ser Ala 15 10
<210> 29 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 3 of HBc18-27 epitope <400> 29
Phe Leu Pro Ala Asp Phe Phe Pro Ser Ile 15 10
<210> 30 <211> 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 4 of HBc18-27 epitope <400> 30
Phe Leu Pro Val Asp Phe Phe Pro Ser Ile 15 10
<210> 31 <211> 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 5 of HBc18-27 epitope <400> 31
Phe Leu Pro Thr Asp Tyr Phe Pro Ser Val 1 5 10
<210> 32 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 6 of HBc18-27 epitope <400> 32
Phe Leu Pro Ser Asp Phe Tyr Pro Pro Val 15 10 <210> 33
<211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 7 of HBc18-27 epitope; 26G <400> 33
Phe Leu Pro Ser Asp Phe Phe Pro Gly Val 15 10
<210> 34 <211> 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 8 of HBc18-27 epitope; 25M, 26G <400> 34
Phe Leu Pro Ser Asp Phe Phe Met Gly Val 15 10
<210> 35 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 9 of HBc18-27 epitope; 23Y <400> 35
Phe Leu Pro Ser Asp Tyr Phe Pro Ser Val 15 10
<210> 36 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 10 of HBc18-27 epitope; 24L <400> 36
Phe Leu Pro Ser Asp Phe Leu Pro Ser Val 15 10
<210> 37 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 11 of HBc18-27 epitope; 21P <400> 37
Phe Leu Pro Pro Asp Phe Phe Pro Ser Val 15 10
<210> 38 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 12 of HBc18-27 epitope; 27A <400> 38
Phe Leu Pro Ser Asp Phe Phe Pro Ser Ala 15 10
<210> 39 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> Natural Variant 13 of HBc18-27 epitope; 24Y <400> 39
Phe Leu Pro Ser Asp Phe Tyr Pro Ser Val 15 10 <210> 40 <211 > 18
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 1 (CDR1) for alpha 12 chain of vwld type TCR <400> 40 gaccgaggtt cccagtcc 18 <210> 41 <211 > 15
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 2 (CDR2) for alpha 12 chain of vuld type TCR <400> 41 afcatactcca atggt 15 <210> 42 <211> 39
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 3 (CDR3) for alpha 12 chain of vwld type TCR <400> 42 tgtgccgtga acctctatgc aggcaacatg ctcaccttt 39 <210> 43 <211>825
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Alpha 12 chain of wild type TCR <400 43 atgatgaaat ccttgagagt tttactagtg atcctgtggc ttcagttgag ctgggtttgg 60 agccaacaga aggaggtgga gcagaattct ggacccctca gtgttccaga gggagccatt 120 gcctctctca actgcactta cagtgaccga ggttcccagt ccttcttctg gtacagacaa 180 tattctggga aaagccctga gttgataatg ttcatatact ccaatggtga caaagaagat 240 ggaaggttta cagcacagct caataaagcc agccagtatg tttctctgcz, catcagagac 300 tcccagccca gtgattcagc cacctacctc tgtgccgtga acctctatgc aggcaacatg 360 ctcacctttg gagggggaac aaggttaatg gtcaaacccc atatccagaa ccctgaccct 420 gccgtgtacc agctgagaga ctctaaatcc agtgacaagt ctgtctgccc attcaccgat 480 tttgattctc aaacaaatgt gtcacaaagt aaggattctg atgtgtatac cacagacaaa 540 actgtgctag acatgaggtc tatggacttc aagagcaaca gtgctgtggc ctggagcaac soo aaatctgact ttgcatgtgc aaacgccttc aacaacagca ttattccaga agacaccttc 660 ttccccagcc cagaaagttc ctgtgatgtc aagctggtcg agaaaagctc tgaaacagat 720 acgaacctaa actttcaaaa cctgtcagtg attgggttcc gaatcctccc cctgaaagtg 780 gccgggttta atctgctcat gacgctgcgg ctgtggtcca gctga 825 <210> 44 <211 > 6
<212> PRT <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 1 (CDR1) for alpha 12 chain of TCR <400> 44
Asp Arg Gly Ser Gin Ser 1 5 <210> 45 <211 > 5
<212> PRT <213> Homo sapiens <220> <221 > Misc_feature <223> Complementarity Determining Region 2 (CDR2) for alpha 12 chain of TCR <400 45 ile Tyr Ser Asn Gly 1 5 <210> 46 <211 > 13
<212> PRT <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 3 (CDR3) for alpha 12 chain of TCR <400> 46
Cys Ala Val Asn Leu Tyr Ala Gly Asn Met Leu Thr Phe 15 10 <210> 47 <211>274
<212> PRT <213> Homo sapiens <220>
<221 > Misc_feature <223> Alpha 12 chain of TCR <400> 47
Met Met Lys Ser Leu Arg Val Leu Leu Val Ile Leu Trp Leu Gin Leu 15 10 15
Ser Trp Val Trp Ser Gin Gin Lys Glu Val Glu Gin Asn Ser Gly Pro 20 25 30
Leu Ser Val Pro Glu Gly Ala Ile Ala Ser Leu Asn Cys Thr Tyr Ser 35 40 45
Asp Arg Gly Ser Gin Ser Phe Phe Trp Tyr Arg Gin Tyr Ser Gly Lys 50 55 60
Ser Pro Glu Leu Ile Met Phe Ile Tyr Ser Asn Gly Asp Lys Glu Asp
65 70 75 BO
Gly Arg Phe Thr Ala Gin Leu Asn Lys Ala Ser Gin Tyr Val Ser Leu 85 90 95
Leu Ile Arg Asp Ser Gin Pro Ser Asp Ser Ala Thr Tyr Leu Cys Ala 100 105 110
Val Asn Leu Tyr Ala Gly Asn Met Leu Thr Phe Gly Gly Gly Thr Arg 115 120 125
Leu Met Val Lys Pro His Ile Gin Asn Pro Asp Pro Ala Val Tyr Gin 130 135 140
Leu Arg Asp Ser Lys Ser Ser Asp Lys Ser Val Cys Leu Phe Thr Asp 145 150 155 160
Phe Asp Ser Gin Thr Asn Val Ser Gin Ser Lys Asp Ser Asp Val Tyr 165 170 175
Ile Thr Asp Lys Thr Val Leu Asp Met Arg Ser Met Asp Phe Lys ser 180 185 190
Asn Ser Ala Val Ala Trp Ser Asn Lys Ser Asp Phe Ala Cys Ala Asn 195 200 205
Ala Phe Asn Asn Ser Ile Ile Pro Glu Asp Thr Phe Phe Pro Ser Pro 210 215 220
Glu Ser Ser Cys Asp Val Lys Leu Val Glu Lys Ser Phe Glu Thr Asp 225 230 235 240
Thr Asn Leu Asn Phe Gin Asn Leu Ser Val Ile Gly Phe Arg ile Leu 245 250 255
Leu Leu Lys Val Ala Gly Phe Asn Leu Leu Met Thr Leu Arg Leu Trp 260 265 270
Ser Ser <210> 48 <211 > 15
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 1 (CDR1) for beta 7.8 chain of vwld type TCR <400 48 togggtcatg tatcc 15 <210> 49 <211 > 18
<212> DNA <213> Homo sapiens <220> <221 > Miscjeature
<223> Complementarity Determining Region 2 (CDR2) for beta 7.8 chain of vwld type TCR <400 49 ttccagaatg aagctcaa 18 <210> 50 <211> 42
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 3 (CDR3) for beta 7.8 chain of vwld type TCR <400=> 50 tgtgccagca gctcggactt tggcaatcag ccccagcatt tt 42 <210 51 <211> 933
<212> DNA <213> Homo sapiens <220> <221 > Misc_feature
<223> Beta 7.8 chain of wild type TCR <400> 51 atgggcacca ggctcctctg atgggtggtc ctgggtttcc tagggacaga tcacacaggt 60 gctggagtct cccagtcccc taggtacaaa gtcgcaaaga gaggacagga tgtagctctc 120 aggtgtgatc caatttcggg tcatgtatcc cttttttggt accaacaggc cctggggcag 180 gggccagagt ttctgactta tttccagaat gaagctcaac tagacaaatc ggggctgccc 240 agtgatcgct tctttgcaga aaggcctgag ggatccgtct ccactctgaa gatccagcgc 300 acacagcagg aggactccgc cgtgtatctc tgtgccagca gctcggactt tggcaatcag 360 ccccagcatt ttggtgatgg gactcgactc tccatcctag aggacctgaa caaggtgttc 420 ccacccgagg tcgctgtgtt tgagccatca gaagcagaga tctcccacac ccaaaaggcc 480 acactggtgt gcctggccac aggcttcttc cctgaccacg tggagctgag ctggtgggtg 540 aatgggaagg aggtgcacag tggggtcagc acggacccgc agcccctcaa ggagcagccc 600 gccctcaatg actccagata ctgcctgagc agccgcctga gggtctcggc caccttctgg 660 cagaaccccc gcaaccactt ccgctgtcaa gtccagttct acgggctctc ggagaatgac 720 gagtggaccc aggatagggc caaacccgtc acccagatcg tcagcgccga ggcctggggt 780 agagcagact gtggctttac ctcggtgtcc taccagcaag gggtcctgtc tgccaccatc 840 ctctatgaga tcctgctagg gaaggccacc ctgtatgctg tgctggtcag cgcccttgtg 900 ttgatggcca tggtcaagag aaaggatttc tga 933 <210> 52 <211 > 5
<212> PRT <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 1 (CDR1) for beta 7.8 chain of TCR <400> 52
Ser Gly His Val Ser 1 5 <210> 53 <211 > 6
<212> PRT <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 2 (CDR2) for beta 7.8 chain of TCR <400> 53
Phe Gin Asn Glu Ala Gin 1 5 <210> 54 <211> 14
<212> PRT <213> Homo sapiens <220> <221 > Misc_feature
<223> Complementarity Determining Region 3 (CDR3) for beta 7.8 chain of TCR <400> 54
Cys Ala Ser Ser Ser Asp Phe Gly Asn Gin Pro Gin His Phe 15 10 <210> 55 <211 > 310
<212> PRT <213> Homo sapiens <220>
<221 > Misc_feature <223> Beta 7.8 chain of TCR <400> 55
Met Gly Thr Arg Leu Leu Cys Trp Val Val Leu Gly Phe Leu Gly Thr 15 10 15
Asp His Thr Gly Ala Gly Val Ser Gin Ser Pro Arg Tyr Lys Val Ala 20 25 30
Lys Arg Gly Gin Asp Val Ala Leu Arg Cys Asp Pro lie Ser Gly His 35 40 45
Val Ser Leu Phe Trp Tyr Gin Gin Ala Leu Gly Gin Gly Pro Glu Phe 50 55 60
Leu Thr Tyr Phe Gin Asn Glu Ala Gin Leu Asp Lys Ser Gly Leu Pro 65 70 75 80
Ser Asp Arg Phe Phe Ala Glu Arg Pro Glu Gly Ser Val Ser Thr Leu 85 90 95
Lys lie Gin Arg Thr Gin Gin Glu Asp Ser Ala Val Tyr Leu Cys Ala 100 105 110
Ser Ser Ser Asp Phe Gly Asn Gin Pro Gin His Phe Gly Asp Gly Thr 115 120 125
Arg Leu Ser lie Leu Glu Asp Leu Asn Lys Val Phe Pro Pro Glu Val 130 135 140
Ala Val Phe Glu Pro Ser Glu Ala Glu lie Ser His Thr Gin Lys Ala 145 150 155 160
Thr Leu Val Cys Leu Ala Thr Gly Phe Phe Pro Asp His Val Glu Leu 165 170 175
Ser Trp Trp Val Asn Gly Lys Glu Val His Ser Gly Val Ser Thr Asp 180 185 190
Pro Gin Pro Leu Lys Glu Gin Pro Ala Leu Asn Asp Ser Arg Tyr Cys 195 200 205
Leu Ser Ser Arg Leu Arg Val Ser Ala Thr phe Trp Gin Asn Pro Arg 210 215 220
Asn His Phe Arg Cys Gin Val Gin Phe Tyr Gly Leu Ser Glu Asn Asp 225 230 235 240
Glu Trp Thr Gin Asp Arg Ala Lys Pro Val Thr Gin lie Val Ser Ala 245 250 255
Glu Ala Trp Gly Arg Ala Asp Cys Gly Phe Thr Ser Val Ser Tyr Gin 260 265 270
Gin Gly Val Leu Ser Ala Thr lie Leu Tyr Glu lie Leu Leu Gly Lys 275 280 285
Ala Thr Leu Tyr Ala Val Leu Val Ser Ala Leu Val Leu Met Ala Met 290 295 300
Val Lys Arg Lys Asp Phe 305 310
<210> 56 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> HBe370-79 epitope (Genotype A/D) <400> 56
Ser Ile Val Ser Pro Phe Ile Pro Leu Leu 15 10
<210> 57 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> HBc18-27 epitope (Genotype B) <220> <221 > Misc_feature <223> HBe370-79 epitope (Genotype B) <400> 57
Asn Ile Leu Ser Pro Phe Met Pro Leu Leu 15 10
<210> 58 <211 > 10 <212> PRT <213> Hepatitis B virus <220> <221 > Misc_feature <223> HBe370-79 epitope (Genotype C) <400> 58
Asn Ile Leu Asn Pro Phe Leu Pro Leu Leu 15 10
REFERENCES CITED IN THE DESCRIPTION
This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.
Non-patent literature cited in the description • J.A. FRELINGERIn immunodominance: The choice of the Immune SvstemWilev-VCH [00591 . KARLINALTSCHULProc. Natl. Acad. Sci., 1990, vol. 87, 2264-68 (00721 . Proc. Natl. Acad. Sci., 1993, vol. 90, 5873-77 TO0721 • SAM BROOKRUSSELLMolecular Cloning: A Laboratory ManualCSHL Press20010000 [0091] [0154] • SAMBROOKRUSSELMolecular Cloning: A Laboratory ManualCold Springs Harbor Laboratory2001000010188] • Current Protocols in ImmunologyJohn Wiley and Sons, lnc.20070000 [01121 [01501 • ENGELS, CAM et al.Kindly provided by Professor Hans StaussRoyal Free and UCL Medical School20030000 (8129] • ENGELS, B.H. CAM et al.Retroviral vectors for high-level transgene expression in T lymphocytesHum Gene Ther, 2003, vol. 14, 121155-6810154] • GEHRING A.J et al.The Level of Viral Antigen Presented by Hepatocytes Influences CD8 T cell FunctionJ Virol., 2007, vol. 81,62940-2949 101541 • KARLIN S.ALTSCHUL S.F Methods for assessing the statistical significance of molecular sequence features by using general scoring schemesProc Natl Acad Sci U S A., 1990, vol. 87, 62264-2268 (0154] • The effect of pathogens on the immune system: Viral hepatitisMAINI, M.BERTOLETTI Ain immunodominance: The choice of the Immune SystemWiley-VCH20060000 (0154] . SCHUMACHERNat Rev. Immunol, 2002, vol. 2, 512- [0154] • STRAIN, A J. et al. Native and recombinant human hepatocyte growth factors are highly potent promoters of DNA synthesis in both human and rat hepatocytesJ Clin Invest, 1991, vol. 87, 1853-7 [0154] • SUN, D.M. NASSALStable HepG2- and Huh7-based human hepatoma cell lines for efficient regulated expression of infectious hepatitis B virusJ Hepatol, 2006, vol. 45, 5636-45 [01541

Claims (14)

HBV-EPITOP-REAKTIV EXOGEN T-CELLERECEPTOR (TCR) OG ANVENDELSER DERAF PATENTKRAVHBV EPITOP-REACTIVE EXOGEN T-CELL RECEPTOR (TCR) AND APPLICATIONS THEREOF 1. Isoleret T-celle, der omfatter mindst én HBV-epitop-reaktiv exogen T-cellereceptor og/eller et fragment deraf, hvor HBV-epitopet er HLA-A2-begrænset, og er: HBV-epitopet er HBcl8-27-epitop, der omfatter SEQ ID NO:25 og/eller SEQ ID NO: 26; og/eller HBs370-79-epitop, der omfatter en hvilken som helst af sekvenserne udvalgt fra gruppen bestående af SEQ ID NO: 56 til 58; og hvor fragmentet omfatter et aktivt sted, der gør det muligt for den HBV-epitop-reaktive exogene T-cellereceptor at genkende HBcl8-27- og/eller HBs370-79-epitopet.An isolated T cell comprising at least one HBV epitope-reactive exogenous T cell receptor and / or a fragment thereof, wherein the HBV epitope is HLA-A2 restricted, and is: The HBV epitope is HBcl8-27 epitope comprising SEQ ID NO: 25 and / or SEQ ID NO: 26; and / or HBs370-79 epitope comprising any of the sequences selected from the group consisting of SEQ ID NOs: 56 to 58; and wherein the fragment comprises an active site which allows the HBV epitope-reactive exogenous T cell receptor to recognize the HBcl8-27 and / or HBs370-79 epitope. 2. Isoleret T-celle ifølge krav 1, hvor den exogene T-cellereceptor omfatter: mindst én α-kæde, der omfatter sekvenser: (a) SEQ ID NO: 9, SEQ ID NO: 10 og SEQ ID NO: 11, og/eller mutanter deraf; og/eller (b) SEQ ID NO: 44, SEQ ID NO: 45 og SEQ ID NO: 46, og/eller mutanter deraf; og/eller mindst én β-kæde, der omfatter sekvenserne: (a) SEQ ID NO: 21, SEQ ID NO: 22 og SEQ ID NO: 23 og/eller mutanter deraf; og/eller (b) SEQ ID NO: 52, SEQ ID NO: 53 og SEQ ID NO: 54 og/eller mutanter deraf, hvor mutanten bibeholder evnen til at binde og aktivere HBV-epitopet.An isolated T cell according to claim 1, wherein the exogenous T cell receptor comprises: at least one α chain comprising sequences: (a) SEQ ID NO: 9, SEQ ID NO: 10 and SEQ ID NO: 11, and / or mutants thereof; and / or (b) SEQ ID NO: 44, SEQ ID NO: 45 and SEQ ID NO: 46, and / or mutants thereof; and / or at least one β chain comprising the sequences: (a) SEQ ID NO: 21, SEQ ID NO: 22 and SEQ ID NO: 23 and / or mutants thereof; and / or (b) SEQ ID NO: 52, SEQ ID NO: 53 and SEQ ID NO: 54, and / or mutants thereof, wherein the mutant retains the ability to bind and activate the HBV epitope. 3. Isoleret T-celle ifølge et hvilket som helst af de foregående krav, hvor den exogene T-cellereceptor omfatter: (a) mindst én α-kæde, der er mindst 90 % aminosyreidentisk med SEQ ID NO: 12 eller et fragment deraf og/eller mindst én β-kæde, der er mindst 90 % aminosyreidentisk med SEQ ID NO: 24 eller et fragment deraf; og/eller (b) mindst én α-kæde, der er mindst 90 % aminosyreidentisk med SEQ ID NO: 47 eller et fragment deraf og/eller mindst én β-kæde, der er mindst 90 % aminosyreidentisk med SEQ ID NO: 55 eller et fragment deraf, hvor fragmentet omfatter et aktivt sted, der gør det muligt for den exogene T-cellereceptor at genkende HBc 18-27- og/eller HBs370-79-epitopet.An isolated T-cell according to any one of the preceding claims, wherein the exogenous T-cell receptor comprises: (a) at least one α-chain which is at least 90% amino acid identical to SEQ ID NO: 12 or a fragment thereof and / or at least one β chain that is at least 90% amino acid identical to SEQ ID NO: 24 or a fragment thereof; and / or (b) at least one α chain that is at least 90% amino acid identical to SEQ ID NO: 47 or a fragment thereof and / or at least one β chain that is at least 90% amino acid identical to SEQ ID NO: 55 or a fragment thereof, wherein the fragment comprises an active site that allows the exogenous T cell receptor to recognize the HBc 18-27 and / or HBs370-79 epitope. 4. Isoleret polynukleotid, der omfatter mindst én sekvens, der koder for mindst én α-kæde og/eller mindst én sekvens, der koder for mindst én β-kæde, hvor a-kæden og β-kæden er af mindst én HBV-epitope-reaktiv exogen T-cellereceptor, hvor HBV-epitopet er HLA-A2-begrænset, og er HBcl8-27-epitopet, der omfatter SEQ ID NO: 25 og/eller SEQ ID NO: 26; og/eller HBs370-79-epitopet, der omfatter en hvilken som helst af sekvenserne udvalgt fra gruppen bestående af SEQ ID NO: 56 til 58.An isolated polynucleotide comprising at least one sequence encoding at least one α chain and / or at least one sequence encoding at least one β chain, wherein the α chain and β chain are of at least one HBV epitope. reactive exogenous T cell receptor wherein the HBV epitope is HLA-A2 restricted and is the HBcl8-27 epitope comprising SEQ ID NO: 25 and / or SEQ ID NO: 26; and / or the HBs370-79 epitope comprising any of the sequences selected from the group consisting of SEQ ID NOs: 56 to 58. 5. Isoleret polynukleotid ifølge krav 4, hvor sekvensen, der koder for α-kæden, omfatter mindst én sekvens udvalgt fra SEQ ID NO: 1 og SEQ ID NO: 5, mindst én sekvens udvalgt fra SEQ ID NO: 2 og SEQ ID NO: 6 og mindst én sekvens udvalgt fra SEQ ID NO: 3 og SEQ TD NO: 7 og/eller sekvensen, der koder for β-kæden, omfatter mindst én sekvens udvalgt fra SEQ ID NO: 13 og SEQ ID NO: 17, mindst én sekvens udvalgt fra SEQ ID NO: 14 og SEQ ID NO: 18 og mindst én sekvens udvalgt fra SEQ ID NO: 15 og SEQ ID NO: 19.An isolated polynucleotide according to claim 4, wherein the sequence encoding the α chain comprises at least one sequence selected from SEQ ID NO: 1 and SEQ ID NO: 5, at least one sequence selected from SEQ ID NO: 2 and SEQ ID NO. : 6 and at least one sequence selected from SEQ ID NO: 3 and SEQ TD NO: 7 and / or the sequence encoding the β chain comprises at least one sequence selected from SEQ ID NO: 13 and SEQ ID NO: 17, at least one sequence selected from SEQ ID NO: 14 and SEQ ID NO: 18 and at least one sequence selected from SEQ ID NO: 15 and SEQ ID NO: 19. 6. Isoleret polynuklcotid ifølge et hvilket som helst af kravene 4 eller 5, hvor sekvensen, der koder for α-kæden, omfatter SEQ ID NO: 5, SEQ ID NO: 6 og SEQ ID NO: 7 og/eller sekvensen, der koder for β-kæden omfatter SEQ ID NO: 17, SEQ ID NO: 18 og SEQ ID NO: 19.An isolated polynucleotide according to any one of claims 4 or 5, wherein the sequence encoding the α chain comprises SEQ ID NO: 5, SEQ ID NO: 6 and SEQ ID NO: 7 and / or the sequence encoding for the β chain, SEQ ID NO: 17 includes SEQ ID NO: 18 and SEQ ID NO: 19. 7. Isoleret polynukleotid ifølge et hvilket som helst af kravene 4 til 6, hvor α-kæden er mindst 90 % sekvensidentisk med SEQ ID NO: 4 eller SEQ ID NO: 8, og β-kæden er mindst 90 % sekvensidentisk med SEQ ID NO: 16 eller SEQ ID NO: 20.An isolated polynucleotide according to any one of claims 4 to 6, wherein the α chain is at least 90% sequence identical to SEQ ID NO: 4 or SEQ ID NO: 8 and the β chain is at least 90% sequence identical to SEQ ID NO. : 16 or SEQ ID NO: 20. 8. Isoleret polynukleotid ifølge krav 4, hvor sekvensen, der koder for α-kæden omfatter SEQ ID NO: 40, SEQ ID NO: 41 og SEQ ID NO: 42 og/eller sekvensen, der koder for β-kæden omfatter SEQ ID NO: 48, SEQ ID NO: 49 og SEQ ID NO: 50.An isolated polynucleotide according to claim 4, wherein the sequence encoding the α chain comprises SEQ ID NO: 40, SEQ ID NO: 41 and SEQ ID NO: 42, and / or the sequence encoding the β chain comprises SEQ ID NO. : 48, SEQ ID NO: 49 and SEQ ID NO: 50. 9. Isoleret polynukleotid ifølge et hvilket som helst af kravene 4 eller 8, hvor α-kæden af den HBV-epitop-reaktive exogen T-ccllcrcccptor cr mindst 90 % sekvensidentisk med SEQ ID NO: 43 og β-kædcn af den HBV-epitop-reaktive exogene T-cellereceptor er mindst 90 % sekvensidentisk med SEQ ID NO: 51.An isolated polynucleotide according to any one of claims 4 or 8, wherein the α chain of the HBV epitope-reactive exogenous T cell receptor is at least 90% sequence identical to SEQ ID NO: 43 and the β chain of the HBV epitope -reactive exogenous T cell receptor is at least 90% sequence identical to SEQ ID NO: 51. 10. Isoleret polypeptid, der omfatter: (a) en α-kæde, der omfatter SEQ ID NO: 9, SEQ ID NO: 10 og SEQ ID NO: 11 og/eller en β-kæde, der omfatter SEQ ID NO: 21, SEQ ID NO: 22 og SEQ ID NO: 23; og/eller (b) en α-kæde, der omfatter SEQ ID NO: 44, SEQ ID NO: 45 og SEQ ID NO: 46 og/eller en β-kæde, der omfatter SEQ ID NO: 52, SEQ ID NO: 53 og SEQ ID NO: 54; hvor polypeptidet er mindst én HBV-epitop-reaktiv exogen T-cellereceptor, og HBV-epitopet er HL A-A2 -begrænset.An isolated polypeptide comprising: (a) an α chain comprising SEQ ID NO: 9, SEQ ID NO: 10 and SEQ ID NO: 11 and / or a β chain comprising SEQ ID NO: 21 , SEQ ID NO: 22 and SEQ ID NO: 23; and / or (b) an α chain comprising SEQ ID NO: 44, SEQ ID NO: 45 and SEQ ID NO: 46, and / or a β chain comprising SEQ ID NO: 52, SEQ ID NO: 53 and SEQ ID NO: 54; wherein the polypeptide is at least one HBV epitope-reactive exogenous T cell receptor and the HBV epitope is HL A-A2 restricted. 11. Isoleret polypeptidet ifølge krav 10, hvor: (a) α-kæden er mindst 90 % amino syreidentisk med SEQ ID NO: 12, og β-kæden er mindst 90 % aminosyreidentisk med SEQ ID NO: 24; og/eller (b) α-kæden er mindst 90% aminosyreidentisk med SEQ ID NO: 47, og β-kæden er mindst 90 % aminosyreidentisk med SEQ ID NO: 55.The isolated polypeptide of claim 10, wherein: (a) the α chain is at least 90% amino acid identical to SEQ ID NO: 12 and the β chain is at least 90% amino acid identical to SEQ ID NO: 24; and / or (b) the α chain is at least 90% amino acid identical to SEQ ID NO: 47 and the β chain is at least 90% amino acid identical to SEQ ID NO: 55. 12. Polypeptid ifølge et hvilket som helst af krav 10 eller 11, hvor polypeptidet er mindst én overvindelig TCR eller et fragment deraf og bundet til mindst ét anti-viralt lægemiddel, hvor fragmentet omfatter et aktivt sted, der gør det muligt for TCR at genkende HBcl8-27- og/eller HBs370-79-epitopet.A polypeptide according to any one of claims 10 or 11, wherein the polypeptide is at least one recoverable TCR or fragment thereof and bound to at least one anti-viral drug, the fragment comprising an active site which allows TCR to recognize The HBcl8-27 and / or HBs370-79 epitope. 13. Isoleret T-celle ifølge et hvilket som helst af kravene 1 til 3, et polynukleotid ifølge et hvilket som helst af kravene 4 til 9 og/eller et polypeptid ifølge et hvilket som helst af kravene 10 til 12 til anvendelse til behandling af en HBV-infektion og/eller et HBV-relateret hepatocellulært karcinom.An isolated T cell according to any one of claims 1 to 3, a polynucleotide according to any one of claims 4 to 9 and / or a polypeptide according to any one of claims 10 to 12 for use in the treatment of a HBV infection and / or an HBV-related hepatocellular carcinoma. 14. Fremgangsmåde in vitro til diagnosticering af mindst ét individ, der er i stand til at overvinde en HBV-infektion, eller som har eller risikerer at få HBV-infektioner og/eller et HBV-relateret hepatocellulært karcinom, hvilken fremgangsmåde omfatter: (a) tilvejebringelse af mindst én prøve fra mindst ét individ; (b) detektering af tilstedeværelsen eller fraværet af mindst ét polynukleotid, der omfatter, i alt væsentligt består af eller består af mindst én nukleinsyresekvens udvalgt fra SEQ ID NO: 4, SEQ ID NO: 8, SEQ ID NO: 16, SEQ ID NO: 20, SEQ ID NO: 43, SEQ ID NO: 51, og/eller en homolog deraf; og/eller (c) detektering af tilstedeværelsen eller fraværet af mindst ét polypeptid, der omfatter, i alt væsentligt består af eller består af aminosyresekvensen SEQ ID NO: 12, SEQ ID NO: 24, SEQ ID NO: 47, SEQ ID NO: 55, og/eller en homolog deraf; hvor tilstedeværelsen af polynukleotidet og/eller polypeptidet er tegn på, at individet er i stand til at overvinde HBV-infektionen og fraværet af polynukleotidet og/eller polypeptidet korrelerer med sandsynligheden for, at individet har eller risikerer at få en HBV-infektion og/eller et HBV-relateret hepatocellulært karcinom.An in vitro method for diagnosing at least one subject capable of overcoming an HBV infection or having or at risk of getting HBV infections and / or an HBV-related hepatocellular carcinoma, comprising: (a) providing at least one sample from at least one individual; (b) detecting the presence or absence of at least one polynucleotide comprising, substantially, or consisting of at least one nucleic acid sequence selected from SEQ ID NO: 4, SEQ ID NO: 8, SEQ ID NO: 16, SEQ ID NO : 20, SEQ ID NO: 43, SEQ ID NO: 51, and / or a homologue thereof; and / or (c) detecting the presence or absence of at least one polypeptide comprising, substantially consisting of or consisting of the amino acid sequence SEQ ID NO: 12, SEQ ID NO: 24, SEQ ID NO: 47, SEQ ID NO: 55, and / or a homologue thereof; wherein the presence of the polynucleotide and / or polypeptide is indicative of the individual being able to overcome the HBV infection and the absence of the polynucleotide and / or polypeptide correlates with the likelihood that the individual has or is at risk of having an HBV infection and / or an HBV-related hepatocellular carcinoma.
DK09742949.2T 2008-05-09 2009-05-07 HBV EPITOP-REACTIVE EXOGEN T-CELL RECEPTOR (TCR) AND APPLICATIONS THEREOF DK2288700T3 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US5197408P 2008-05-09 2008-05-09
PCT/SG2009/000165 WO2009136874A1 (en) 2008-05-09 2009-05-07 Hbv epitope reactive exogenous t cell receptor (tcr) and uses thereof

Publications (1)

Publication Number Publication Date
DK2288700T3 true DK2288700T3 (en) 2017-05-22

Family

ID=41264797

Family Applications (1)

Application Number Title Priority Date Filing Date
DK09742949.2T DK2288700T3 (en) 2008-05-09 2009-05-07 HBV EPITOP-REACTIVE EXOGEN T-CELL RECEPTOR (TCR) AND APPLICATIONS THEREOF

Country Status (6)

Country Link
US (3) US8603810B2 (en)
EP (1) EP2288700B1 (en)
CN (1) CN102083979B (en)
DK (1) DK2288700T3 (en)
ES (1) ES2622505T3 (en)
WO (1) WO2009136874A1 (en)

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2805564A1 (en) * 2010-08-05 2012-02-09 Stefan Jenewein Anti-mhc antibody anti-viral cytokine fusion protein
EP3333186B9 (en) 2012-03-28 2023-10-04 Gadeta B.V. Combinatorial gamma 9 delta 2 t cell receptor chain exchange
US20140004079A1 (en) * 2012-06-21 2014-01-02 Agency For Science, Technology And Research In vivo dendritic cell therapeutic adjuvant
GB201313377D0 (en) * 2013-07-26 2013-09-11 Adaptimmune Ltd T cell receptors
CA2968457C (en) * 2014-11-20 2023-12-05 Umc Utrecht Holding B.V. Use of antibodies for enrichment of engineered t cells with exogenous immune receptors and antibodies for use in depletion of engineered t cells
CN108463548B (en) * 2015-10-30 2023-04-18 加利福尼亚大学董事会 Method for producing T cell from stem cell and immunotherapy method using the T cell
US11020431B2 (en) 2016-01-06 2021-06-01 Health Research, Inc. Compositions and libraries comprising recombinant T-cell receptors and methods of using recombinant T-cell receptors
WO2017171631A1 (en) 2016-03-31 2017-10-05 Lion Tcr Pte. Ltd. Non-activated t cells expressing exogenous virus-specific t cell receptor (tcr)
CA3027124A1 (en) 2016-06-10 2017-12-14 Umc Utrecht Holding B.V. Novel method for identifying deltat-cell (or gammat-cell) receptor chains or parts thereof that mediate an anti-tumour or an anti-infective response
JP7038703B2 (en) * 2016-09-23 2022-03-18 ライオン ティーシーアール ピーティーイー.エルティーディー. HBV antigen-specific binding molecule and fragments thereof
DE102016123893A1 (en) 2016-12-08 2018-06-14 Immatics Biotechnologies Gmbh T cell receptors with improved binding
CN108948184B (en) * 2017-05-22 2021-04-23 香雪生命科学技术(广东)有限公司 T cell receptor for recognizing PRAME antigen-derived short peptide
CN107827959B (en) * 2017-11-09 2018-10-30 杭州续缓生物科技有限公司 Identify the TCR and application thereof of hepatitis B (HBV) surface antigen S 183-91 epitopes
CN110156889B (en) * 2018-02-14 2023-03-10 中国科学院广州生物医药与健康研究院 High affinity HBs T cell receptor
WO2020010565A1 (en) * 2018-07-12 2020-01-16 深圳华大生命科学研究院 Mart-1(27-35) epitope-specific t cell receptor
US20230087348A1 (en) * 2019-01-11 2023-03-23 Agency For Science, Technology And Research Use of hla-a*11:01-restricted hepatitis b virus (hbv) peptides for identifying hbv-specific cd8+ t cells
JP2022521513A (en) * 2019-02-20 2022-04-08 フレッド ハッチンソン キャンサー リサーチ センター RAS neoantigen-specific binding proteins and their use
CN112940105B (en) * 2019-11-26 2022-08-16 中国科学院微生物研究所 HLA-A11 restrictive hepatitis B virus HBc 141-151 T cell receptor of epitope peptide and application thereof
WO2021148110A1 (en) * 2020-01-21 2021-07-29 Lion Tcr Pte. Ltd. Hbv specific tcr library and its use as personalised medicine
CN111592589B (en) * 2020-05-22 2022-05-10 深圳市因诺转化医学研究院 Specific TCR for recognizing human hepatitis B virus core antigen C18-27 epitope
CN111592590B (en) * 2020-05-22 2022-05-03 深圳市因诺转化医学研究院 T cell receptor recognizing human hepatitis B virus core antigen
TWI815194B (en) 2020-10-22 2023-09-11 美商基利科學股份有限公司 INTERLEUKIN-2-Fc FUSION PROTEINS AND METHODS OF USE
GB202109807D0 (en) * 2021-07-07 2021-08-18 Ucl Business Ltd Cell
CN117003856B (en) * 2023-09-27 2023-12-01 南方医科大学南方医院 T cell receptor engineering T cell of targeting hepatitis B surface antigen

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6322789B1 (en) * 1991-08-26 2001-11-27 Epimmune, Inc. HLA-restricted hepatitis B virus CTL epitopes
US6235288B1 (en) * 1992-08-26 2001-05-22 The Scripps Research Institute Peptides for inducing cytotoxic T lymphocyte responses to hepatitis B virus
CA2352738A1 (en) * 1998-12-04 2000-06-08 Biogen, Inc. Hbv core antigen particles with multiple immunogenic components attached via peptide ligands
CA2580412A1 (en) * 2004-09-13 2006-03-23 Government Of The United States Of America, Represented By The Secretary , Department Of Health And Human Services Compositions comprising t cell receptors and methods of use thereof
WO2007034489A2 (en) * 2005-09-22 2007-03-29 Yeda Res & Dev Immunogenic fragments of t-cell receptor constant domains and peptides derived therefrom
US8119772B2 (en) * 2006-09-29 2012-02-21 California Institute Of Technology MART-1 T cell receptors
CN101036784B (en) * 2007-03-09 2010-11-10 中国人民解放军第二军医大学 Toxicity T cell position vaccine of the cell for treating Hepatitis B and the preparing method

Also Published As

Publication number Publication date
CN102083979B (en) 2015-01-07
US10004801B2 (en) 2018-06-26
EP2288700B1 (en) 2017-02-08
US20110070208A1 (en) 2011-03-24
US20140134194A1 (en) 2014-05-15
EP2288700A4 (en) 2012-10-10
US8603810B2 (en) 2013-12-10
WO2009136874A1 (en) 2009-11-12
CN102083979A (en) 2011-06-01
ES2622505T3 (en) 2017-07-06
US20170202953A1 (en) 2017-07-20
EP2288700A1 (en) 2011-03-02

Similar Documents

Publication Publication Date Title
DK2288700T3 (en) HBV EPITOP-REACTIVE EXOGEN T-CELL RECEPTOR (TCR) AND APPLICATIONS THEREOF
JP7184645B2 (en) Modified Chimeric Receptors and Related Compositions and Methods
JP6180114B2 (en) T cell receptor
EP3310805B1 (en) Pd-1-cd28 fusion proteins and their use in medicine
CN107827959B (en) Identify the TCR and application thereof of hepatitis B (HBV) surface antigen S 183-91 epitopes
EP3099708B1 (en) Methods and compositions for producing a cell expressing a t cell receptor
JP2020517308A (en) TCR and peptides
JP2019525898A (en) Human leukocyte antigen-restricted gamma delta T cell receptor and method of use thereof
WO2016095783A1 (en) T cell receptor for identifying eb virus short peptide
US20130217122A1 (en) Expansion of Interferon-Gamma-Producing T-Cells Using Glypican-3 Peptide Library
AU2017205637A1 (en) Compositions and libraries comprising recombinant T-cell receptors and methods of using recombinant T-cell receptors
JP2019533435A (en) HBV antigen-specific binding molecules and fragments thereof
WO2020089433A2 (en) Tcr and peptides
WO2023239290A1 (en) Hbv surface antigen specific t cell receptors and uses thereof
JP2023518446A (en) Compositions and methods for treating lupus
EP4067371A1 (en) T-cell receptors specific for hepatitis c virus peptides
US20240009235A1 (en) T cell receptors directed against bob1 and uses thereof
JP2022153316A (en) T-cell receptor
Barbaro et al. A New Strategy of Tumor Vaccination Based on Mammary Adenocarcinoma Cells Transduced with the MHC Class II Transactivator CIITA. L. Mortara1, P. Castellani2, R. Meazza3, G. Tosi1, A. De